EP3538123A1 - Activin receptor type iia variants and methods of use thereof - Google Patents

Activin receptor type iia variants and methods of use thereof

Info

Publication number
EP3538123A1
EP3538123A1 EP17868622.6A EP17868622A EP3538123A1 EP 3538123 A1 EP3538123 A1 EP 3538123A1 EP 17868622 A EP17868622 A EP 17868622A EP 3538123 A1 EP3538123 A1 EP 3538123A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
seq
subject
bone
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP17868622.6A
Other languages
German (de)
French (fr)
Other versions
EP3538123A4 (en
Inventor
Jasbir S. Seehra
Jennifer Lachey
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Keros Therapeutics Inc
Original Assignee
Keros Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Keros Therapeutics Inc filed Critical Keros Therapeutics Inc
Publication of EP3538123A1 publication Critical patent/EP3538123A1/en
Publication of EP3538123A4 publication Critical patent/EP3538123A4/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/1103Receptor protein serine/threonine kinase (2.7.11.30)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • Healthy bone undergoes a constant remodeling that involves both bone breakdown and bone growth. Bone growth is mediated by the osteoblast cell type whereas the osteoclasts resorb the bone Pathology occurs when these systems fall out of balance either through downregulation of the anabolic program, upregulation of the catabolic system or a combination of both, resulting in a net bone loss. Therefore, controlling the balance in bone remodeling can be useful for promoting the healing of fractures and other damage to bone as well as the treatment of disorders, such as osteoporosis, associated with loss of bone mass and bone mineralization.
  • Bone damage can result from a range of root causes, including age- or cancer-related bone loss, genetic conditions, adverse side effects of drug treatment, or fracture.
  • the World Health Organization estimates that osteoporosis alone affects 75 million people in the U.S., Europe and Japan, and is a significant risk factor in bone fracture. In general, the whole of bone loss represents pathological states for which there are few effective treatments. Treatment instead focuses on immobilization, exercise and dietary modifications rather than agents that directly promote bone growth and increase bone density. With respect to osteoporosis, estrogen, calcitonin, osteocalcin with vitamin K, or high doses of dietary calcium are all used as therapeutic interventions.
  • the present invention features polypeptides that include an extracellular activin receptor type lla
  • a polypeptide of the invention includes an extracellular ActRlla variant fused to the N- or C-terminus of an Fc domain monomer or moiety. Such moieties may be attached by amino acid or other covalent bonds.
  • a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may also form a dimer (e.g., a homodimer or heterodimer) through the interaction between two Fc domain monomers.
  • the polypeptides of the invention may be used to increase bone mass or bone mineral density in a subject having a disease or condition involving bone damage, e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility.
  • a disease or condition involving bone damage e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility.
  • polypeptides of the invention may also be used to affect myostatin, activin, and/or bone morphogenetic protein 9 (BMP9) signaling in a subject having a risk of developing or having a disease or condition involving bone damage or bone demineralization.
  • BMP9 bone morphogenetic protein 9
  • the invention features a polypeptide including an extracellular activin receptor type lla (ActRlla) variant, the variant having a sequence of
  • the variant has a sequence of
  • GAILG RSETQECLFX2NANWX3X4X5X6TNQTGVEX7CX8GX9KX11 X12X13X14HCX15ATWX16NISGSIEIVX17X1 8GCX19X20X21 DX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 2), wherein X2, X3, X4, Xs, ⁇ , X7, Xs, X9, Xn , X12, X13, X14, X15, X16, X17, Xis, X19, X20, X21 , X22, X23, X24, X25, X26, and X27 are defined as above.
  • the variant has a sequence of
  • GAILG RSETQECLFX2NANWEX4X5RTNQTGVEX7CX8GX9KDKRX14HCX15ATWX16NISGSI EIVKX18GCWL DDX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 3), wherein X2, X4, Xs, X7, Xs, X9, X14, X15, X16, Xis, X22, X23, X24, X25, X26, and X27 are defined as above.
  • the variant has a sequence of
  • GAILG RSETQECLFX2NANWEX 4 DRTNQTGVEX7CXsGX9KDKRXi4HCXi5ATWX 16 NISGSIEIVKXi8GCWL DDX22NCYDRTDCVEX23KX25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 4), wherein X2, X4, X7, Xs, X9, X14, X15, X 16 , Xis, X22, X23, X25, X26, and X27 are defined as above.
  • the variant has a sequence of
  • Xi is F or Y.
  • X2 is F or Y.
  • X3 is E or A.
  • X4 is K or L.
  • X5 is D or E.
  • is R or A.
  • X7 is P or R.
  • Xs is Y or E.
  • X9 is D or E.
  • X10 is K or Q.
  • Xn is D or A.
  • X12 is K or A.
  • X13 is R or A.
  • Xi4 is R or L.
  • X15 is F or Y.
  • X16 is K, R, or A.
  • X17 is K, A, Y, F, or I.
  • Xis is Q or K.
  • X19 is W or A.
  • X20 is L or A.
  • X21 is D, K, R, A, F, G, M, N, or I.
  • X22 is I, F, or A.
  • X23 is K or T.
  • X24 is K or E.
  • X25 is D or E.
  • X26 is S or N.
  • X27 is E or Q.
  • X23 is T
  • X24 is E
  • X25 is E
  • X26 is N
  • X23 is T
  • X24 is K
  • X25 is E
  • X26 is N
  • X17 is K.
  • the variant has the sequence of any one of SEQ ID NOs: 6-72.
  • the amino acid at position X24 may be replaced with the amino acid K.
  • the amino acid at position X24 may be replaced with the amino acid E.
  • a polypeptide described herein may further include a C-terminal extension of one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6 more amino acids).
  • the C-terminal extension is amino acid sequence NP.
  • the C- terminal extension is amino acid sequence NPVTPK (SEQ ID NO: 155).
  • a polypeptide described herein may further include a moiety fused or covalently linked to the C-terminus of the polypeptide.
  • the moiety may increase the stability of improve pharmacokinetic properties of the polypeptide.
  • the moiety is an Fc domain monomer, an Fc domain, an albumin binding peptide, a fibronectin domain, or serum albumin.
  • a polypeptide described herein may further include an Fc domain monomer fused to the C-terminus of the polypeptide by way of a linker.
  • the polypeptide that includes an extracellular ActRMa variant described herein fused to an Fc domain monomer may form a dimer (e.g., a homodimer or heterodimer) through the interaction between two Fc domain monomers.
  • the Fc domain monomer has the sequence of SEQ ID NO: 97
  • a polypeptide described herein may further include an Fc domain fused to the C-terminus of the polypeptide by way of a linker.
  • the Fc domain is a wild-type Fc domain.
  • the wild-type Fc domain has the sequence of SEQ ID NO: 151 .
  • the Fc domain contains one or more amino acid substitutions.
  • the Fc domain containing one or more amino acid substitutions does not form a dimer.
  • a polypeptide described herein may further include an albumin-binding peptide fused to the C-terminus of the polypeptide by way of a linker.
  • the albumin-binding peptide has the sequence of SEQ ID NO: 1 52.
  • a polypeptide described herein may further include a fibronectin domain fused to the C-terminus of the polypeptide by way of a linker.
  • the fibronectin domain peptide has the sequence of SEQ ID NO: 153.
  • a polypeptide described herein may further include a human serum albumin fused to the C-terminus of the polypeptide by way of a linker.
  • the human serum albumin has the sequence of SEQ ID NO: 154.
  • the linker is an amino acid spacer.
  • the amino acid spacer is GGG, GGGA (SEQ ID NO: 98), GGGG (SEQ ID NO: 1 00), GGGAG (SEQ ID NO: 130), GGGAGG (SEQ ID NO: 131 ), or GGGAGGG (SEQ ID NO: 132).
  • the amino acid spacer is GGGS (SEQ ID NO: 99), GGGGA (SEQ ID NO: 101 ), GGGGS (SEQ ID NO: 102), GGGGG (SEQ ID NO: 103), GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), AGGG (SEQ ID NO: 106), SGGG (SEQ ID NO: 107), GAGA (SEQ ID NO: 108), GSGS (SEQ ID NO: 109), GAGAGA (SEQ ID NO: 1 10), GSGSGS (SEQ ID NO: 1 1 1 ), GAGAGAGA (SEQ ID NO: 1 12), GSGSGSGS (SEQ ID NO: 1 13), GAGAGAGAGA (SEQ ID NO: 1 14), GSGSGSGSGS (SEQ ID NO: 1 15), GAGAGAGAGA (SEQ ID NO: 1 16), and GSGSGSGSGSGSGS (SEQ ID NO: 1 1 7), GGAGGA (SEQ ID NO:
  • GGGGSGGGGSGGGGS SEQ ID NO: 129
  • AAAL SEQ ID NO: 133
  • AAAK SEQ ID NO: 134
  • AAAR SEQ ID NO: 135)
  • EGKSSGSGSESKST SEQ ID NO: 136
  • G SAG S A AG SG E F SEQ ID NO: 137
  • AEAAAKEAAAKA SEQ ID NO: 138
  • KESGSVSSEQLAQFRSLD SEQ ID NO: 139
  • GENLYFQSGG SEQ ID NO: 140
  • SACYCELS SEQ ID NO: 141
  • RSIAT SEQ ID NO: 142
  • RPACKIPNDLKQKVMNH SEQ ID NO: 143
  • AAANSSIDLISVPVDSR (SEQ ID NO: 145), GGSGGGSEGGGSEGGGSEGGGSEGGGSEGGGSGGGS (SEQ ID NO: 146), EAAAK (SEQ ID NO: 147), or PAPAP(SEQ ID NO: 148).
  • the polypeptide described herein has a serum half- life of at least 7 days.
  • the polypeptide described herein binds to human bone morphogenetic protein 9 (BMP9) with a KD of 200 pM or higher. In some embodiments, the polypeptide binds to activin and/or myostatin and has reduced (e.g., weak) binding to human BMP9. In some embodiments, the polypeptide does not substantially bind to human BMP9.
  • BMP9 bone morphogenetic protein 9
  • the polypeptide described herein binds to human activin A with a KD of 800 pM or less.
  • the polypeptide described herein binds to human activin B with a KD of approximately 800 pM or less.
  • the polypeptide described herein binds to human GDF-1 1 with a KD of approximately 5 pM or higher.
  • the invention features a nucleic acid molecule encoding a polypeptide described herein (e.g., a polypeptide including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)).
  • a polypeptide described herein e.g., a polypeptide including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • the invention also features a vector including the nucleic acid molecule described herein.
  • the invention features a host cell that expresses a polypeptide described herein, wherein the host cell includes a nucleic acid molecule or a vector described in the previous two aspects, wherein the nucleic acid molecule or vector is expressed in the host cell.
  • the invention features a method of preparing a polypeptide described herein, wherein the method includes: a) providing a host cell including a nucleic acid molecule or a vector described herein, and b) expressing the nucleic acid molecule or vector in the host cell under conditions that allow for the formation of the polypeptide.
  • the invention features a pharmaceutical composition including a polypeptide, nucleic acid molecule, or vector described herein and one or more pharmaceutically acceptable carriers or excipients.
  • the polypeptide, nucleic acid molecule, or vector is in a therapeutically effective amount.
  • the invention also features a construct including two identical polypeptides
  • an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to the N- or C-terminus of an Fc domain monomer (e.g., the sequence of SEQ ID NO: 97).
  • the two Fc domain monomers in the two polypeptides interact to form an Fc domain in the construct.
  • the invention also features a construct including two different polypeptides
  • a heterodimer each including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to the N- or C-terminus of an Fc domain monomer.
  • the two Fc domain monomers in the two polypeptides interact to form an Fc domain in the construct.
  • the invention features a method of increasing bone mineral density in a subject in need thereof.
  • the method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of reducing bone resorption in a subject in need thereof.
  • the method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of increasing bone formation in a subject in need thereof.
  • the method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the subject has primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.
  • the invention features a method of affecting myostatin, activin, and/or BMP9 signaling (e.g., reducing or inhibiting the binding of myostatin, activin, and/or BMP9 to their receptors) in a subject having a disease or condition involving bone damage, wherein method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the disease or condition is primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment- related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity- related bone loss, or immobility-related bone loss.
  • the invention features a method of treating a subject having primary osteoporosis by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having secondary osteoporosis by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having osteopenia by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having a fracture by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having bone cancer or cancer metastasis-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having Paget's disease by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject renal osteodystrophy by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having treatment-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having diet-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having bone loss associated with the treatment of obesity by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having low gravity-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the invention features a method of treating a subject having immobility-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
  • the primary osteoporosis is age-related osteoporosis. In some embodiments of any of the above aspects, the primary osteoporosis is hormone-related osteoporosis.
  • the secondary osteoporosis is immobilization- induced osteoporosis.
  • the secondary osteoporosis is glucocorticoid-induced osteoporosis.
  • the cancer is multiple myeloma.
  • the treatment is FGF-21 treatment.
  • the treatment is GLP-1 treatment.
  • the treatment is cancer therapy.
  • the treatment is treatment for obesity and/or Type-2 diabetes.
  • the diet-related bone loss is rickets.
  • the method increases bone formation in the subject. In some embodiments of any of the above aspects, wherein the method decreases bone resorption in the subject. In some embodiments of any of the above aspects, the method increases osteoblast activity or osteoblastogenesis. In some embodiments of any of the above aspects, the method decreases osteoclast activity or decreases osteoclastogenesis. In some embodiments of any of the above aspects, the method reduces or inhibits the binding of activin and/or myostatin to their receptors.
  • the method does not cause a vascular complication (e.g., an increase vascular permeability or leakage) in the subject. In some embodiments of any of the methods described herein, the method increases bone mineral density in the subject.
  • a vascular complication e.g., an increase vascular permeability or leakage
  • the bone is cortical bone. In some embodiments of any of the above aspects, the bone is trabecular bone.
  • the polypeptide, nucleic acid, vector, or pharmaceutical composition is administered in an amount sufficient to increase bone density, reduce bone resorption, reduce the rate of bone resorption, increase bone formation, increase the rate of bone formation, reduce osteoclast activity, increase osteoblast activity, or affect myostatin, activin, and/or BMP9 signaling in the subject.
  • the variant has the sequence of SEQ ID NO: 69.
  • the variant having the sequence of SEQ ID NO: 69 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 58.
  • the variant having the sequence of SEQ ID NO: 58 has the amino acid K at position X17, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • a C- terminal extension e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 6.
  • the variant having the sequence of SEQ ID NO: 6 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 38.
  • the variant having the sequence of SEQ ID NO: 38 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss
  • the variant has the sequence of SEQ ID NO: 41 .
  • the variant having the sequence of SEQ ID NO: 41 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 44.
  • the variant having the sequence of SEQ ID NO: 44 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 70.
  • the variant having the sequence of SEQ ID NO: 70 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 71 .
  • the variant having the sequence of SEQ ID NO: 71 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids VTPK).
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-
  • the variant has the sequence of SEQ ID NO: 72.
  • the variant having the sequence of SEQ ID NO: 72 has the amino acid K at position Xi7 and/or the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26.
  • the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary
  • osteoporosis osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss
  • Paget's disease, renal osteodystrophy treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss
  • myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss
  • Paget's disease, renal osteodystrophy treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 72, optionally having the amino acid K at position X17 and/or the amino acid sequence TEEN or TKEN
  • extracellular activin receptor type Ma (ActRlla) variant refers to a peptide including a soluble, extracellular portion of the single transmembrane receptor, ActRlla, that has at least one amino acid substitution relative to a wild-type extracellular ActRlla (e.g., bold portion of the sequence of SEQ ID NO: 75 shown below) or an extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96.
  • the sequence of the wild-type, human ActRlla precursor protein is shown below (SEQ ID NO: 75), in which the signal peptide is italicized and the extracellular portion is bold. Wild-type, human ActRlla precursor protein (SEQ ID NO: 75):
  • An extracellular ActRlla variant may have a sequence of any one of SEQ ID NOs: 1 -72.
  • an extracellular ActRlla variant has a sequence of any one of SEQ ID NOs: 6-72 (Table 2).
  • an extracellular ActRlla variant may have at least 85% (e.g., at least 85%, 87%, 90%, 92%, 95%, 97%, or greater) amino acid sequence identity to the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73).
  • extracellular ActRllb variant refers to a peptide including a soluble, extracellular portion of the single transmembrane receptor, ActRllb, that has at least one amino acid substitution relative to a wild-type extracellular ActRllb (e.g., the sequence of SEQ ID NO: 74).
  • An extracellular ActRllb variant may have the sequence of SEQ ID NO: 149 shown below:
  • linker refers to a linkage between two elements, e.g., peptides or protein domains.
  • a polypeptide described herein may include an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)) fused to a moiety.
  • the moiety may increase stability or improve pharmacokinetic properties of the polypeptide.
  • the moiety may be fused to the polypeptide by way of a linker.
  • a linker can be a covalent bond or a spacer.
  • the term "bond" refers to a chemical bond, e.g., an amide bond or a disulfide bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation.
  • spacer refers to a moiety (e.g., a polyethylene glycol (PEG) polymer) or an amino acid sequence (e.g., a 1 -200 amino acid sequence) occurring between two elements, e.g., peptides or protein domains, to provide space and/or flexibility between the two elements.
  • An amino acid spacer is part of the primary sequence of a polypeptide (e.g., fused to the spaced peptides via the polypeptide backbone).
  • the formation of disulfide bonds, e.g., between two hinge regions that form an Fc domain is not considered a linker.
  • Fc domain refers to a dimer of two Fc domain monomers.
  • An Fc domain has at least 80% sequence identity (e.g., at least 85%, 90%, 95%, 97%, or 100% sequence identity) to a human Fc domain that includes at least a CH2 domain and a CH3 domain.
  • An Fc domain monomer includes second and third antibody constant domains (CH2 and CH3).
  • the Fc domain monomer also includes a hinge domain.
  • An Fc domain does not include any portion of an immunoglobulin that is capable of acting as an antigen-recognition region, e.g., a variable domain or a complementarity determining region (CDR).
  • CDR complementarity determining region
  • each of the Fc domain monomers in an Fc domain includes amino acid substitutions in the CH2 antibody constant domain to reduce the interaction or binding between the Fc domain and an Fey receptor.
  • the Fc domain contains one or more amino acid substitutions that reduce or inhibit Fc domain dimerization.
  • An Fc domain can be any immunoglobulin antibody isotype, including IgG, IgE, IgM, IgA, or IgD. Additionally, an Fc domain can be an IgG subtype (e.g., lgG1 , lgG2a, lgG2b, lgG3, or lgG4). The Fc domain can also be a non-naturally occurring Fc domain, e.g., a recombinant Fc domain.
  • albumin-binding peptide refers to an amino acid sequence of 12 to 16 amino acids that has affinity for and functions to bind serum albumin.
  • An albumin-binding peptide can be of different origins, e.g., human, mouse, or rat.
  • an albumin-binding peptide has the sequence DICLPRWGCLW (SEQ ID NO: 1 52).
  • fibronectin domain refers to a high molecular weight glycoprotein of the extracellular matrix, or a fragment thereof, that binds to, e.g., membrane-spanning receptor proteins such as integrins and extracellular matrix components such as collagens and fibrins.
  • a fibronectin domain is a fibronectin type III domain (SEQ ID NO: 153) having amino acids 61 0-702 of the sequence of UniProt ID NO: P02751 .
  • a fibronectin domain is an adnectin protein.
  • human serum albumin refers to the albumin protein present in human blood plasma. Human serum albumin is the most abundant protein in the blood. It constitutes about half of the blood serum protein. In some embodiments, a human serum albumin has the sequence of UniProt ID NO: P02768 (SEQ ID NO: 154).
  • fused is used to describe the combination or attachment of two or more elements, components, or protein domains, e.g., peptides or polypeptides, by means including chemical conjugation, recombinant means, and chemical bonds, e.g., amide bonds.
  • two single peptides in tandem series can be fused to form one contiguous protein structure, e.g., a polypeptide, through chemical conjugation, a chemical bond, a peptide linker, or any other means of covalent linkage.
  • an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused in tandem series to the N- or C-terminus of a moiety (e.g., Fc domain monomer (e.g., the sequence of SEQ ID NO: 97) a wild-type Fc domain (e.g., the sequence of SEQ ID NO: 151 ), an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide (e.g., the sequence of SEQ ID NO: 152), a fibronectin domain (e.g., the sequence of SEQ ID NO: 1 53), or a human serum albumin (e.g., the sequence of SEQ ID NO: 154)) by way of a linker.
  • a moiety e.g., Fc domain mono
  • an extracellular ActRlla variant is fused to a moiety (e.g., an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin) by way of a peptide linker, in which the N-terminus of the peptide linker is fused to the C- terminus of the extracellular ActRlla variant through a chemical bond, e.g., a peptide bond, and the C- terminus of the peptide linker is fused to the N-terminus of the moiety (e.g., Fc domain monomer, wild- type Fc domain, Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), albumin-binding peptide, fibronectin domain, or human serum albumin) through a chemical bond, e.
  • C-terminal extension refers to the addition of one or more amino acids to the C-terminus of a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)).
  • the C-terminal extension can be one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, or more amino acids).
  • Exemplary C- terminal extensions are the amino acid sequence NP (a two amino acid C-terminal extension) and the amino acid sequence NPVTPK (SEQ ID NO: 1 55) (a six amino acid C-terminal extension).
  • SEQ ID NO: 71 which is the sequence of SEQ ID NO: 69 with a C-terminal extension of NP
  • SEQ ID NO: 72 which is the sequence of SEQ ID NO: 69 with a C-terminal extension of NPVTPK
  • percent (%) identity refers to the percentage of amino acid (or nucleic acid) residues of a candidate sequence, e.g., an extracellular ActRlla variant, that are identical to the amino acid (or nucleic acid) residues of a reference sequence, e.g., a wild-type extracellular ActRlla (e.g., SEQ ID NO: 73), after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity (i.e., gaps can be introduced in one or both of the candidate and reference sequences for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the percent amino acid (or nucleic acid) sequence identity of a given candidate sequence to, with, or against a given reference sequence is calculated as follows:
  • A is the number of amino acid (or nucleic acid) residues scored as identical in the alignment of the candidate sequence and the reference sequence
  • B is the total number of amino acid (or nucleic acid) residues in the reference sequence.
  • the percent amino acid (or nucleic acid) sequence identity of the candidate sequence to the reference sequence would not equal to the percent amino acid (or nucleic acid) sequence identity of the reference sequence to the candidate sequence.
  • a reference sequence aligned for comparison with a candidate sequence may show that the candidate sequence exhibits from 50% to 100% identity across the full length of the candidate sequence or a selected portion of contiguous amino acid (or nucleic acid) residues of the candidate sequence.
  • the length of the candidate sequence aligned for comparison purpose is at least 30%, e.g., at least 40%, e.g., at least 50%, 60%, 70%, 80%, 90%, or 100% of the length of the reference sequence.
  • a position in the candidate sequence is occupied by the same amino acid (or nucleic acid) residue as the corresponding position in the reference sequence, then the molecules are identical at that position.
  • the term "serum half-life" refers to, in the context of administering a therapeutic protein to a subject, the time required for plasma concentration of the protein in the subject to be reduced by half.
  • the protein can be redistributed or cleared from the bloodstream, or degraded, e.g., by proteolysis.
  • a polypeptide including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)
  • binding affinity refers to the strength of the binding interaction between two molecules.
  • binding affinity refers to the strength of the sum total of non-covalent interactions between a molecule and its binding partner, such as an extracellular ActRlla variant and BMP9 or activin A.
  • binding affinity refers to intrinsic binding affinity, which reflects a 1 :1 interaction between members of a binding pair.
  • the binding affinity between two molecules is commonly described by the dissociation constant (KD) or the affinity constant (KA). TWO molecules that have low binding affinity for each other generally bind slowly, tend to dissociate easily, and exhibit a large KD.
  • TWO molecules that have high affinity for each other generally bind readily, tend to remain bound longer, and exhibit a small KD.
  • the KD of two interacting molecules may be determined using methods and techniques well known in the art, e.g., surface plasmon resonance. KD is calculated as the ratio of k 0 ff/k on .
  • bone mineral density (BMD)
  • bone density and “bone mass” refer to a measure of the amount of bone mineral (e.g. calcium) in bone tissue.
  • BMD may be measured by well- established clinical techniques known to one of skill in the art (e.g., by single-1 or dual-energy photon or X-ray absorptiometry).
  • the concept of BMD relates to the mass of mineral per volume of bone, although clinically it is measured by proxy according to optical density per square centimeter of bone surface upon imaging. BMD measurement is used in clinical medicine as an indirect indicator of osteoporosis and fracture risk.
  • BMD test results are provided as a T-score, where the T-score represents the BMD of a subject compared to the ideal or peak bone mineral density of a healthy 30-year- old adult.
  • a score of 0 indicates that the BMD is equal to the normal reference value for a healthy young adult.
  • Differences between the measured BMD of subject and that of the reference value for a healthy young adult are measured in standard deviations units (SDs).
  • SDs standard deviations units
  • a T-score of between +1 SD and -1 SD may indicate a normal BMD
  • a T-score of between -1 SD and -2.5 SD may indicate low bone mass (e.g., osteopenia)
  • a T-score lower than -2.5 SD may indicate osteoporosis or severe osteoporosis.
  • a polypeptide of the invention including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, wherein the patient has low bone mass (e.g., a T-Score of between -1 SD and -2.5 SD).
  • a polypeptide of the invention including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, wherein the patient has osteoporosis (e.g., a T-Score of less than -2.5 SD).
  • administration of a polypeptide of the invention including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide or a vector containing such a nucleic acid molecule treats the subject by increasing their BMD.
  • administration of a polypeptide of the invention including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule increases the BMD of a subject resulting in an increase in the T- Score of the subject (e.g., resulting in an increase in the T-Score of the subject of 0.1 or more, 0.2 or more, 0.3 or more, 0.4 or more, 0.5 or more, 1 .0 or more, or 2.0 or more).
  • bone remodeling or “bone metabolism” refer to the process for maintaining bone strength and ion homeostasis by replacing discrete parts of old bone with newly synthesized packets of proteinaceous matrix. Bone is resorbed by osteoclasts, and is deposited by osteoblasts in a process called ossification. Osteocyte activity plays a key role in this process.
  • Conditions that result in a decrease in bone mass can either be caused by an increase in resorption, or a decrease in ossification.
  • bone formation exceeds resorption.
  • resorption exceeds formation.
  • Bone resorption rates are also typically much higher in post-menopausal older women due to estrogen deficiency related to menopause.
  • bone resorption or “bone catabolic activity” refer to a process by which osteoclasts break down the tissue in bones and release the minerals, resulting in a transfer of the mineral (e.g., calcium) from bone tissue to the blood.
  • Increased rates of bone resorption are associated with aging, including in post-menopausal women.
  • High rates of bone resorption, or rates of bone resorption that exceed the rate of ossification, are associated with bone disorders, such as decreased bone mineral density, including osteopenia and osteoporosis.
  • a polypeptide of the invention including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof to decrease done resorption in the subject (e.g., the rate of bone resorption in the subject).
  • a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, to increase bone formation (e.g., increase the rate of bone formation or osteogenesis in the subject).
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, to increase bone formation (e.g., increase the rate of bone formation or osteogenesis in the subject).
  • affecting myostatin, activin, and/or BMP9 signaling means changing the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla).
  • a polypeptide including an extracellular ActRlla variant described herein reduces or inhibits the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla).
  • a polypeptide of the invention including an extracellular ActRlla variant may have weak binding affinity to BMP9 (e.g., KD of 200 pM or higher).
  • vascular complication refers to a vascular disorder or any damage to the blood vessels, such as damage to the blood vessel walls. Damage to the blood vessel walls may cause an increase in vascular permeability or leakage.
  • vascular permeability or leakage refers to the capacity of the blood vessel walls to allow the flow of small molecules, proteins, and cells in and out of blood vessels.
  • An increase in vascular permeability or leakage may be caused by an increase in the gaps (e.g., an increase in the size and/or number of the gaps) between endothelial cells that line the blood vessel walls and/or thinning of the blood vessel walls.
  • polypeptide describes a single polymer in which the monomers are amino acid residues which are covalently conjugated together through amide bonds.
  • a polypeptide is intended to encompass any amino acid sequence, either naturally occurring, recombinant, or synthetically produced.
  • the term "homodimer” refers to a molecular construct formed by two identical macromolecules, such as proteins or nucleic acids.
  • the two identical monomers may form a homodimer by covalent bonds or non-covalent bonds.
  • an Fc domain may be a homodimer of two Fc domain monomers if the two Fc domain monomers contain the same sequence.
  • a polypeptide described herein including an extracellular ActRlla variant fused to an Fc domain monomer may form a homodimer through the interaction of two Fc domain monomers, which form an Fc domain in the homodimer.
  • heterodimer refers to a molecular construct formed by two different macromolecules, such as proteins or nucleic acids.
  • the two monomers may form a heterodimer by covalent bonds or non-covalent bonds.
  • a polypeptide described herein including an extracellular ActRlla variant fused to an Fc domain monomer may form a heterodimer through the interaction of two Fc domain monomers, each fused to a different ActRlla variant, which form an Fc domain in the heterodimer.
  • the term "host cell” refers to a vehicle that includes the necessary cellular components, e.g., organelles, needed to express proteins from their corresponding nucleic acids.
  • the nucleic acids are typically included in nucleic acid vectors that can be introduced into the host cell by conventional techniques known in the art (transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, etc.).
  • a host cell may be a prokaryotic cell, e.g., a bacterial cell, or a eukaryotic cell, e.g., a mammalian cell (e.g., a CHO cell or a HEK293 cell).
  • the term "therapeutically effective amount” refers an amount of a polypeptide, nucleic acid, or vector of the invention or a pharmaceutical composition containing a polypeptide, nucleic acid, or vector of the invention effective in achieving the desired therapeutic effect in treating a patient having a disease, such as -osteoporosis, or a condition involving bone damage, e.g., primary
  • therapeutically effective amount also refers an amount of a polypeptide, nucleic acid, or vector of the invention or a pharmaceutical composition containing a polypeptide, nucleic acid, or vector of the invention effective in achieving the desired therapeutic effect in treating a patient having a disease, such as a disease or condition involving bone damage.
  • the therapeutically effective amount of the polypeptide, nucleic acid, or vector avoids adverse side effects.
  • the term "pharmaceutical composition” refers to a medicinal or pharmaceutical formulation that includes an active ingredient as well as excipients and diluents to enable the active ingredient suitable for the method of administration.
  • the pharmaceutical composition of the present invention includes pharmaceutically acceptable components that are compatible with the polypeptide, nucleic acid, or vector.
  • the pharmaceutical composition may be in tablet or capsule form for oral administration or in aqueous form for intravenous or subcutaneous administration.
  • the term "pharmaceutically acceptable carrier or excipient” refers to an excipient or diluent in a pharmaceutical composition.
  • the pharmaceutically acceptable carrier must be compatible with the other ingredients of the formulation and not deleterious to the recipient.
  • the pharmaceutically acceptable carrier or excipient must provide adequate pharmaceutical stability to the polypeptide including an extracellular ActRlla variant, the nucleic acid molecule(s) encoding the polypeptide, or a vector containing such nucleic acid molecule(s).
  • the nature of the carrier or excipient differs with the mode of administration. For example, for intravenous administration, an aqueous solution carrier is generally used; for oral administration, a solid carrier is preferred.
  • treating and/or preventing refers to the treatment and/or prevention of a disease, e.g., a bone disease or condition (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), using methods and compositions of the invention.
  • a bone disease or condition e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss
  • treating a bone disease or condition occurs after a subject has developed
  • Preventing a bone disease or condition refers to steps or procedures taken when a subject is at risk of developing the bone disease or condition.
  • the subject may show signs or mild symptoms that are judged by a physician to be indications or risk factors for developing the bone disease or condition or have a family history or genetic predisposition of developing the bone disease or condition, but has not yet developed the disease.
  • the term "subject" refers to a mammal, e.g., preferably a human. Mammals include, but are not limited to, humans and domestic and farm animals, such as monkeys (e.g., a cynomolgus monkey), mice, dogs, cats, horses, and cows, etc.
  • monkeys e.g., a cynomolgus monkey
  • mice dogs, cats, horses, and cows, etc.
  • FIG. 1 is a sequence alignment showing the wild-type sequences of extracellular ActRlla and ActRllb and the amino acid substitutions in ActRlla variants. DETAILED DESCRIPTION OF THE INVENTION
  • a polypeptide of the invention includes an extracellular ActRlla variant fused to a moiety (e.g., Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin).
  • a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may also form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers.
  • the ActRlla variants described herein have weak binding affinity or no binding affinity to bone morphogenetic protein 9 (BMP9) compared to activins and myostatin.
  • the invention also includes methods of treating diseases and conditions involving bone damage by increasing bone mineral density or bone formation or affecting myostatin, activin, and/or BMP9 signaling in a subject by administering to the subject a polypeptide including an extracellular ActRlla variant described herein.
  • Activin type II receptors are single transmembrane domain receptors that modulate signals for ligands in the transforming growth factor ⁇ (TGF- ⁇ ) superfamily.
  • Ligands in the TGF- ⁇ superfamily are involved in a host of physiological processes, such as muscle growth, vascular growth, cell differentiation, homeostasis, and osteogenesis.
  • Examples of ligands in the TGF- ⁇ superfamily include, e.g., activin, inhibin, growth differentiation factors (GDFs) (e.g., GDF8, also known as myostatin), and bone morphogenetic proteins (BMPs) (e.g., BMP9).
  • GDFs growth differentiation factors
  • BMPs bone morphogenetic proteins
  • Activin has been reported to be upregulated in bone disease and inhibit osteoblast activity. Myostatin is also implicated in bone homeostasis through increasing osteogenesis and inhibiting osteoblast activity.
  • activin receptor ligands e.g., activin and myostatin
  • promote bone resorption which could lead to diseases and conditions involving bone damage, such as primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.
  • Methods that reduce or inhibit this signaling could, therefore, be used in the treatment of diseases and conditions involving bone damage.
  • ActRlla activin type II receptors
  • ActRllb activin type II receptors
  • Studies have shown that BMP9 binds ActRllb with about 300-fold higher binding affinity than ActRlla (see, e.g., Townson et al., J. Biol. Chem. 287:27313, 2012).
  • ActRlla is known to have a longer half-life compared to ActRllb.
  • the present invention describes extracellular ActRlla variants that are constructed by introducing amino acid residues of ActRllb to ActRlla, with the goal of imparting physiological properties conferred by ActRllb, while also maintaining beneficial physiological and pharmacokinetic properties of ActRlla.
  • the optimum peptides confer significant increases in bone mineral density, while retaining longer serum half-life and low binding-affinity to BMP9, for example.
  • the preferred ActRlla variants also exhibit improved binding to activins and/or myostatin compared to wild-type ActRlla, which allows them to compete with endogenous activin receptors for ligand binding and reduce or inhibit endogenous activin receptor signaling. These variants can be used to treat disorders in which activin receptor signaling is elevated, such bone disease, leading to a reduction in bone resorption or osteoclast activity, and in increase in bone formation, bone mineral density, or bone strength.
  • amino acid substitutions may be introduced to an extracellular ActRlla variant to reduce or remove the binding affinity of the variant to BMP9.
  • the wild- type amino acid sequences of the extracellular portions of human ActRlla and ActRllb are shown below.
  • Polypeptides described herein include an extracellular ActRlla variant having at least one amino acid substitution relative to the wild-type extracellular ActRlla having the sequence of SEQ ID NO: 73 or the extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96. Possible amino acid substitutions at 27 different positions may be introduced to an extracellular ActRlla variant (Table 1 ).
  • an extracellular ActRlla variant may have at least 85% (e.g., at least 85%, 87%, 90%, 92%, 95%, 97%, or greater) amino acid sequence identity to the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73).
  • An extracellular ActRlla variant may have one or more (e.g., 1 -27, 1 -25, 1 -23, 1 -21 , 1 -19, 1 -17, 1 -15, 1 -13, 1 -1 1 , 1 -9, 1 -7, 1 -5, 1 -3, or 1 -2; e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 1 7, 18, 19, 20, 21 , 22, 23, 24, 25, 26, or 27) amino acid substitutions relative the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73).
  • an extracellular ActRlla variant may include amino acid substitutions at all of the 27 positions as listed in Table 1 .
  • an extracellular ActRlla variant may include amino acid substitutions at a number of positions, e.g., at 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, or 26 out of the 27 positions, as listed in Table 1 .
  • Amino acid substitutions can worsen or improve the activity and/or binding affinity of the ActRlla variants of the invention.
  • the lysine (K) at position Xi7 in the sequences shown in Tables 1 and 2 SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) be retained. Substitutions at that position can lead to a loss of activity.
  • RTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 150) has reduced activity in vivo, indicating that the substitution of alanine (A) for lysine (K) at X17 is not tolerated.
  • ActRlla variants of the invention including variants in Tables 1 and 2 (e.g., SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72), therefore, retain amino acid K at position X17.
  • the ActRlla variants of the invention preferably have reduced, weak, or no substantial binding to
  • BMP9 binding is reduced in ActRlla variants containing the amino acid sequence TEEN at positions X23, X24, X25, and X26, as well as in variants that maintain the amino acid K at position X24 and have the amino acid sequence TKEN at positions X23, X24, X25, and X26.
  • the sequences TEEN and TKEN can be employed interchangeably in the ActRlla variants (e.g., the variants in Tables 1 and 2, e.g., SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) of the invention to provide reduced BMP9 binding.
  • the ActRlla variants of the invention may further include a C-terminal extension (e.g., additional amino acids at the C-terminus).
  • the C-terminal extension can add one or more additional amino acids at the C-terminus (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids) to any of the variants shown in Tables 1 and 2 (e.g., SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)).
  • One potential C-terminal extension that can be included in the ActRlla variants of the invention is amino acid sequence NP.
  • the sequence including the C-terminal extension is SEQ ID NO: 71 (e.g., SEQ ID NO: 69 with a C- terminal extension of NP).
  • Another exemplary C-terminal extension that can be included in the ActRlla variants of the invention is amino acid sequence NPVTPK (SEQ ID NO: 155).
  • the sequence including the C-terminal extension is SEQ ID NO: 72 (e.g., SEQ ID NO: 69 with a C-terminal extension of NPVTPK).
  • X 3 is E, X 6 is R, Xi i is D, X12 is K, X13 is R, Xi 6 is K or R, X17 is K, X19 is W, X20 is L, X21 is D, and X22 is I or F.
  • Xi7 is K.
  • Xi7 is K, X23 is T, X24 is E, X25 is E, and X26 is N.
  • X17 is K
  • X23 is T
  • X24 is K
  • X25 is E
  • X26 is N.
  • a polypeptide described herein includes an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 6-72 (Table 2).
  • a polypeptide of the invention including an extracellular ActRlla variant (e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) has amino acid K at position X17. Altering the amino acid at position X17 can result in reduced activity.
  • GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIVAKGCWLDDFNCYD RTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 150) has reduced activity in vivo, indicating that the substitution of A for K at X17 is not tolerated.
  • a polypeptide of the invention including an extracellular ActRlla variant e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) with the sequence TEEN at positions X23, X24, X25, and X26 can have a substitution of the amino acid K for the amino acid E at position X24.
  • a polypeptide of the invention including an extracellular ActRlla variant e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • the sequence TKEN at positions X23, X24, X25, and X26 can have a substitution of the amino acid E for the amino acid K at position X24.
  • Polypeptides having the sequence TEEN or TKEN at positions X23, X24, X25, and X26 have reduced or weak binding to BMP9.
  • a polypeptide of the invention including an extracellular ActRlla variant may further include a C-terminal extension (e.g., additional amino acids at the C-terminus).
  • the C-terminal extension is amino acid sequence NP.
  • the sequence including the C-terminal extension is SEQ ID NO: 71 (e.g., SEQ ID NO: 69 with a C-terminal extension of NP).
  • the C-terminal extension is amino acid sequence NPVTPK (SEQ ID NO: 155).
  • the sequence including the C-terminal extension is SEQ ID NO: 72 (e.g., SEQ ID NO: 69 with a C-terminal extension of NPVTPK).
  • the C-terminal extension can add one or more additional amino acids at the C-terminus (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids).
  • a polypeptide of the invention including an extracellular ActRlla variant may further include a moiety (e.g., Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin), which may be fused to the N- or C-terminus (e.g., C- terminus) of the extracellular ActRlla variant by way of a linker.
  • the moiety increases the stability or improves the pharmacokinetic properties of the polypeptide.
  • a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers, which combine to form an Fc domain in the dimer.
  • a dimer e.g., homodimer or heterodimer
  • an extracellular ActRlla variant described herein does not have the sequence of any one of SEQ ID NOs: 76-96 shown in Table 3 below.
  • a polypeptide described herein has a serum half-life of at least 7 days in humans.
  • the polypeptide may bind to bone morphogenetic protein 9 (BMP9) with a KD of 200 pM or higher.
  • BMP9 bone morphogenetic protein 9
  • the polypeptide may bind to activin A with a KD of 10 pM or higher.
  • the polypeptide does not bind to BMP9 or activin A.
  • the polypeptide binds to activin and/or myostatin and exhibits reduced (e.g., weak) binding to BMP9.
  • the polypeptide that has reduced or weak binding to BMP9 has the sequence TEEN or TKEN at positions X23, X24, X25, and X26.
  • the polypeptide may bind to human BMP9 with a KD of about 200 pM or higher (e.g., a KD of about 200, 300, 400, 500, 600, 700, 800, or 900 pM or higher, e.g., a KD of about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, or 50 nM or higher, e.g., a KD of between about 200 pM and about 50 nM).
  • the polypeptide does not substantially bind to human BMP9.
  • the polypeptide may bind to human activin A with a KD of about 800 pM or less (e.g., a KD of about 800, 700, 600, 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 pM or less, e.g., a KD of between about 800 pM and about 200 pM).
  • a KD of about 800 pM or less e.g., a KD of about 800, 700, 600, 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 pM or less, e.g., a KD of between about 800 pM and about 200 pM.
  • the polypeptide may bind to human activin B with a KD of 800 pM or less (e.g., a KD of about 800, 700, 600, 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 pM or less, e.g., a KD of between about 800 pM and about 200 pM)
  • the polypeptide may also bind to growth and differentiation factor 1 1 (GDF-1 1 ) with a KD of approximately 5 pM or higher (e.g., a KD of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 1 05, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 1 70, 175, 180, 185, 190, 195, or 200 pM or higher).
  • GDF-1 1 growth and
  • a polypeptide described herein may include an extracellular ActRlla variant fused to an Fc domain monomer of an immunoglobulin or a fragment of an Fc domain to increase the serum half-life of the polypeptide.
  • a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers, which form an Fc domain in the dimer.
  • an Fc domain is the protein structure that is found at the C-terminus of an immunoglobulin.
  • An Fc domain includes two Fc domain monomers that are dimerized by the interaction between the CH3 antibody constant domains.
  • a wild-type Fc domain forms the minimum structure that binds to an Fc receptor, e.g., FcyRI, FcyRlla, FcyRllb, FcyRllla, FcyRlllb, FcyRIV.
  • an Fc domain may be mutated to lack effector functions, typical of a "dead" Fc domain.
  • an Fc domain may include specific amino acid substitutions that are known to minimize the interaction between the Fc domain and an Fey receptor.
  • an Fc domain is from an lgG1 antibody and includes amino acid substitutions L234A, L235A, and G237A.
  • an Fc domain is from an lgG1 antibody and includes amino acid substitutions D265A, K322A, and N434A.
  • the aforementioned amino acid positions are defined according to Kabat (Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1 991 )).
  • the Kabat numbering of amino acid residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a "standard" Kabat numbered sequence.
  • an Fc domain does not induce any immune system-related response.
  • the Fc domain in a dimer of a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may be modified to reduce the interaction or binding between the Fc domain and an Fey receptor.
  • the sequence of an Fc domain monomer that may be fused to an extracellular ActRlla variant is shown below (SEQ ID NO: 97):
  • an Fc domain is from an lgG1 antibody and includes amino acid substitutions L12A, L13A, and G15A, relative to the sequence of SEQ ID NO: 97. In some embodiments, an Fc domain is from an lgG1 antibody and includes amino acid substitutions D43A, K100A, and N212A, relative to the sequence of SEQ ID NO: 97.
  • an extracellular ActRlla variant described herein may be fused to the N- or C-terminus of an Fc domain monomer (e.g., SEQ ID NO: 97) through conventional genetic or chemical means, e.g., chemical conjugation.
  • a linker e.g., a spacer
  • the extracellular ActRlla variant and the Fc domain monomer may be fused to the extracellular ActRlla variant and the Fc domain monomer.
  • the Fc domain monomer can be fused to the N- or C-terminus (e.g., C-terminus) of the extracellular ActRlla variant.
  • a polypeptide described herein may include an extracellular ActRlla variant fused to an Fc domain.
  • the Fc domain contains one or more amino acid substitutions that reduce or inhibit Fc domain dimerization.
  • the Fc domain contains a hinge domain.
  • the Fc domain can be of immunoglobulin antibody isotype IgG, IgE, IgM, IgA, or IgD.
  • the Fc domain can be an IgG subtype (e.g., lgG1 , lgG2a, lgG2b, lgG3, or lgG4).
  • the Fc domain can also be a non-naturally occurring Fc domain, e.g., a recombinant Fc domain.
  • one or more amino acids with large side-chains may be introduced to the CH3-CH3 dimer interface to hinder dimer formation due to steric clash.
  • one or more amino acids with small side-chains e.g., alanine, valine, or threonine
  • Methods of introducing amino acids with large or small side-chains in the CH3 domain are described in, e.g., Ying et al. (J Biol Chem.
  • one or more amino acid residues in the CH3 domain that make up the CH3-CH3 interface between two Fc domains are replaced with positively-charged amino acid residues (e.g., lysine, arginine, or histidine) or negatively-charged amino acid residues (e.g., aspartic acid or glutamic acid) such that the interaction becomes electrostatically unfavorable depending on the specific charged amino acids introduced.
  • positively-charged amino acid residues e.g., lysine, arginine, or histidine
  • negatively-charged amino acid residues e.g., aspartic acid or glutamic acid
  • an Fc domain includes one or more of the following amino acid substitutions:T366W, T366Y, T394W, F405W, Y349T, Y349E, Y349V, L351 T, L351 H, L351 N, L352K, P353S, S354D, D356K, D356R, D356S, E357K, E357R, E357Q, S364A, T366E, L368T, L368Y, L368E, K370E, K370D, K370Q, K392E, K392D, T394N, P395N, P396T, V397T, V397Q, L398T, D399K, D399R, D399N, F405T, F405H, F405R, Y407T, Y407H, Y407I, K409E, K409
  • a polypeptide described herein may include an extracellular ActRlla variant fused to a serum protein-binding peptide. Binding to serum protein peptides can improve the pharmacokinetics of protein pharmaceuticals.
  • albumin-binding peptides that can be used in the methods and compositions described here are generally known in the art.
  • the albumin binding peptide includes the sequence DICLPRWGCLW (SEQ ID NO: 1 52).
  • albumin-binding peptides may be joined to the N- or C-terminus (e.g., C- terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant.
  • an albumin-binding peptide is joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
  • an extracellular ActRlla variant described herein may be fused to the N- or C-terminus of albumin-binding peptide (e.g., SEQ ID NO: 152) through conventional genetic or chemical means, e.g., chemical conjugation.
  • a linker e.g., a spacer
  • inclusion of an albumin-binding peptide in an extracellular ActRlla variant described herein may lead to prolonged retention of the therapeutic protein through its binding to serum albumin.
  • a polypeptide described herein may include an extracellular ActRlla variant fused to fibronectin domains. Binding to fibronectin domains can improve the pharmacokinetics of protein pharmaceuticals.
  • Fibronectin domain is a high molecular weight glycoprotein of the extracellular matrix, or a fragment thereof, that binds to, e.g., membrane-spanning receptor proteins such as integrins and extracellular matrix components such as collagens and fibrins.
  • a fibronectin domain is joined to the N- or C-terminus (e.g., C-terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant.
  • a fibronectin domain can be joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
  • fibronectin domains that can be used in the methods and compositions described here are generally known in the art.
  • the fibronectin domain is a fibronectin type III domain (SEQ ID NO: 153) having amino acids 610-702 of the sequence of UniProt ID NO:
  • the fibronectin domain is an adnectin protein.
  • an extracellular ActRlla variant described herein may be fused to the N- or C-terminus of a fibronectin domain (e.g., SEQ ID NO: 153) through conventional genetic or chemical means, e.g., chemical conjugation.
  • a linker e.g., a spacer
  • a linker can be inserted between the extracellular ActRlla variant and the fibronectin domain.
  • a polypeptide described herein may include an extracellular ActRlla variant fused to serum albumin. Binding to serum albumins can improve the pharmacokinetics of protein pharmaceuticals.
  • Serum albumin is a globular protein that is the most abundant blood protein in mammals. Serum albumin is produced in the liver and constitutes about half of the blood serum proteins. It is monomeric and soluble in the blood. Some of the most crucial functions of serum albumin include transporting hormones, fatty acids, and other proteins in the body, buffering pH, and maintaining osmotic pressure needed for proper distribution of bodily fluids between blood vessels and body tissues. In preferred embodiments, serum albumin is human serum albumin.
  • a human serum albumin is joined to the N- or C-terminus (e.g., C-terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant.
  • an extracellular ActRlla variant described herein e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a human serum albumin can be joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
  • serum albumins that can be used in the methods and compositions described herein are generally known in the art.
  • the serum albumin includes the sequence of UniProt ID NO: P02768 (SEQ ID NO: 154).
  • an extracellular ActRlla variant described herein may be fused to the N- or C-terminus of a human serum albumin (e.g., SEQ ID NO: 1 54) through conventional genetic or chemical means, e.g., chemical conjugation.
  • a linker e.g., a spacer
  • inclusion of a serum albumin in an extracellular ActRlla variant described herein may lead to prolonged retention of the therapeutic protein.
  • a polypeptide described herein may include an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety by way of a linker.
  • the moiety increases stability of the polypeptide.
  • moieties include an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin.
  • a linker between a moiety e.g., an Fc domain monomer (e.g., the sequence of SEQ ID NO: 97), a wild-type Fc domain (e.g., SEQ ID NO: 151 ), an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce
  • an albumin-binding peptide e.g., SEQ ID NO: 152
  • a fibronectin domain e.g., SEQ ID NO: 153
  • a human serum albumin e.g., SEQ ID NO: 1 54
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)
  • Suitable peptide spacers are known in the art, and include, for example, peptide linkers containing flexible amino acid residues such as glycine, alanine, and serine.
  • a spacer can contain motifs, e.g., multiple or repeating motifs, of GA, GS, GG, GGA, GGS, GGG, GGGA (SEQ ID NO: 98), GGGS (SEQ ID NO: 99), GGGG (SEQ ID NO: 100), GGGGA (SEQ ID NO: 101 ), GGGGS (SEQ ID NO: 102), GGGGG (SEQ ID NO: 103), GGAG (SEQ ID NO: 1 04), GGSG (SEQ ID NO: 105), AGGG (SEQ ID NO: 1 06), or SGGG (SEQ ID NO: 107).
  • motifs e.g., multiple or repeating motifs, of GA, GS, GG, GGA, GGS, GGG, GGGA (SEQ ID NO: 98), GGGS (SEQ ID NO: 99), GGGG (SEQ ID NO: 100), GGGGA (SEQ ID NO: 101 ), GGGGS (SEQ ID NO: 102), GGGGG
  • a spacer can contain 2 to 12 amino acids including motifs of GA or GS, e.g., GA, GS, GAGA (SEQ ID NO: 1 08), GSGS (SEQ ID NO: 109), GAGAGA (SEQ ID NO: 1 1 0), GSGSGS (SEQ ID NO: 1 1 1 ), GAGAGAGA (SEQ ID NO: 1 12), GSGSGSGS (SEQ ID NO: 1 13),
  • a spacer can contain 3 to 12 amino acids including motifs of GGA or GGS, e.g., GGA, GGS, GGAGGA (SEQ ID NO: 1 1 8), GGSGGS (SEQ ID NO: 1 19), GGAGGAGGA (SEQ ID NO: 120), GGSGGSGGS (SEQ ID NO: 121 ), GGAGGAGGAGGA (SEQ ID NO: 122), and GGSGGSGGSGGS (SEQ ID NO: 123).
  • a spacer can contain 4 to 12 amino acids including motifs of GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), e.g., GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), GGAGGGAG (SEQ ID NO: 124), GGSGGGSG (SEQ ID NO: 125), GGAGGGAGGGAG (SEQ ID NO: 126), and
  • a spacer can contain motifs of GGGGA (SEQ ID NO: 101 ) or GGGGS (SEQ ID NO: 102), e.g., GGGGAGGGGAGGGGA (SEQ ID NO: 128) and GGGGSGGGGSGGGGS (SEQ ID NO: 129).
  • an amino acid spacer between a moiety e.g., an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a spacer can also contain amino acids other than glycine, alanine, and serine, e.g., AAAL (SEQ ID NO: 133), AAAK (SEQ ID NO: 134), AAAR (SEQ ID NO: 135),
  • EGKSSGSGSESKST SEQ ID NO: 136
  • GSAGSAAGSGEF SEQ ID NO: 137
  • AEAAAKEAAAKA SEQ ID NO: 138
  • KESGSVSSEQLAQFRSLD SEQ ID NO: 139
  • G ENLYFQSGG SEQ ID NO: 140
  • SACYCELS SEQ ID NO: 141
  • RSIAT SEQ ID NO: 142
  • RPACKIPNDLKQKVMNH SEQ ID NO: 143
  • AAANSSIDLISVPVDSR SEQ ID NO: 145
  • GGSGGGSEGGGSEGGGSEGGGSEGGGSEGGGSGGGSGGGS SEQ ID NO: 146
  • a spacer can contain motifs, e.g., multiple or repeating motifs, of EAAAK (SEQ ID NO: 147). In some embodiments, a spacer can contain motifs, e.g., multiple or repeating motifs, of proline-rich sequences such as (XP)n, in which X may be any amino acid (e.g., A, K, or E) and n is from 1 - 5, and PAPAP(SEQ ID NO: 148).
  • XP proline-rich sequences
  • the length of the peptide spacer and the amino acids used can be adjusted depending on the two protein involved and the degree of flexibility desired in the final protein fusion polypeptide.
  • the length of the spacer can be adjusted to ensure proper protein folding and avoid aggregate formation.
  • the polypeptides of the invention can be produced from a host cell.
  • a host cell refers to a vehicle that includes the necessary cellular components, e.g., organelles, needed to express the polypeptides and fusion polypeptides described herein from their corresponding nucleic acids.
  • the nucleic acids may be included in nucleic acid vectors that can be introduced into the host cell by conventional techniques known in the art (e.g., transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, infection, or the like).
  • transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, infection, or the like The choice of nucleic acid vectors depends in part on the host cells to be used. Generally, preferred host cells are of either eukaryotic (e.g., mammalian) or prokaryotic (e.g., bacterial) origin.
  • a nucleic acid sequence encoding the amino acid sequence of a polypeptide of the invention may be prepared by a variety of methods known in the art. These methods include, but are not limited to, oligonucleotide-mediated (or site-directed) mutagenesis and PCR mutagenesis.
  • a nucleic acid molecule encoding a polypeptide of the invention may be obtained using standard techniques, e.g., gene synthesis.
  • a nucleic acid molecule encoding a wild-type extracellular ActRlla may be mutated to include specific amino acid substitutions using standard techniques in the art, e.g., QuikChangeTM mutagenesis.
  • Nucleic acid molecules can be synthesized using a nucleotide synthesizer or PCR techniques.
  • a nucleic acid sequence encoding a polypeptide of the invention may be inserted into a vector capable of replicating and expressing the nucleic acid molecule in prokaryotic or eukaryotic host cells.
  • Many vectors are available in the art and can be used for the purpose of the invention.
  • Each vector may include various components that may be adjusted and optimized for compatibility with the particular host cell.
  • the vector components may include, but are not limited to, an origin of replication, a selection marker gene, a promoter, a ribosome binding site, a signal sequence, the nucleic acid sequence encoding protein of interest, and a transcription termination sequence.
  • mammalian cells may be used as host cells for the invention.
  • mammalian cell types include, but are not limited to, human embryonic kidney (HEK) (e.g., HEK293, HEK 293F), Chinese hamster ovary (CHO), HeLa, COS, PC3, Vera, MC3T3, NSO, Sp2/0, VERY, BHK, MDCK, W138, BT483, Hs578T, HTB2, BT20, T47D, NSO (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7030, and HsS78Bst cells.
  • HEK human embryonic kidney
  • CHO Chinese hamster ovary
  • HeLa HeLa
  • COS e.g., COS, PC3, Vera
  • MC3T3, NSO a murine myeloma cell line that does not endogenously produce any immunoglobulin chains
  • CRL7030 a murine my
  • E. coli strains include, but are not limited to, E. coli 294 (ATCC ® 31 ,446), E. coli K 1776 (ATCC ® 31 ,537, E. coli BL21 (DE3) (ATCC ® BAA- 1025), and E. coli RV308 (ATCC ® 31 ,608).
  • E. coli 294 ATCC ® 31 ,446
  • E. coli K 1776 ATCC ® 31 ,537
  • E. coli BL21 DE3
  • E. coli RV308 ATCC ® 31 ,608.
  • Different host cells have characteristic and specific mechanisms for the posttranslational processing and modification of protein products (e.g., glycosylation). Appropriate cell lines or host systems may be chosen to ensure the correct modification and processing of the polypeptide expressed.
  • the above-described expression vectors may be introduced into appropriate host cells using conventional techniques in the art, e.g., transformation, transfection, electroporation, calcium phosphate precipitation, and direct microinjection.
  • host cells are cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • Methods for expression of therapeutic proteins are known in the art, see, for example, Paulina Balbas, Argelia Lorence (eds.) Recombinant Gene Expression: Reviews and Protocols (Methods in Molecular Biology), Humana Press; 2nd ed. 2004 and Vladimir Voynov and Justin A. Caravella (eds.) Therapeutic Proteins: Methods and Protocols (Methods in Molecular Biology) Humana Press; 2nd ed. 2012.
  • Host cells used to produce the polypeptides of the invention may be grown in media known in the art and suitable for culturing of the selected host cells.
  • suitable media for mammalian host cells include Minimal Essential Medium (MEM), Dulbecco's Modified Eagle's Medium (DMEM), Expi293TM Expression Medium, DMEM with supplemented fetal bovine serum (FBS), and RPMI-1 640.
  • suitable media for bacterial host cells include Luria broth (LB) plus necessary supplements, such as a selection agent, e.g., ampicillin.
  • Host cells are cultured at suitable temperatures, such as from about 20 °C to about 39 °C, e.g., from 25 °C to about 37 °C, preferably 37 °C, and C0 2 levels, such as 5 to 10%.
  • the pH of the medium is generally from about 6.8 to 7.4, e.g., 7.0, depending mainly on the host organism. If an inducible promoter is used in the expression vector of the invention, protein expression is induced under conditions suitable for the activation of the promoter.
  • the expressed protein may be secreted from the host cells (e.g., mammalian host cells) into the cell culture media. Protein recovery may involve filtering the cell culture media to remove cell debris.
  • the proteins may be further purified.
  • a polypeptide of the invention may be purified by any method known in the art of protein purification, for example, by chromatography (e.g., ion exchange, affinity, and size-exclusion column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • the protein can be isolated and purified by appropriately selecting and combining affinity columns such as Protein A column (e.g., POROS Protein A chromatography) with chromatography columns (e.g., POROS HS-50 cation exchange chromatography), filtration, ultra filtration, salting-out and dialysis procedures.
  • affinity columns such as Protein A column (e.g., POROS Protein A chromatography) with chromatography columns (e.g., POROS HS-50 cation exchange chromatography), filtration, ultra filtration, salting-out and dialysis procedures.
  • host cells may be disrupted, e.g., by osmotic shock, sonication, or lysis, to recover the expressed protein. Once the cells are disrupted, cell debris may be removed by osmotic shock, sonication, or lysis.
  • a polypeptide can be conjugated to marker sequences, such as a peptide to facilitate purification.
  • marker amino acid sequence is a hexa- histidine peptide (His-tag), which binds to nickel-functionalized agarose affinity column with micromolar affinity.
  • His-tag hexa- histidine peptide
  • Other peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA" tag, which corresponds to an epitope derived from influenza hemagglutinin protein (Wilson et al., Cell 37:767, 1984).
  • the polypeptides of the invention can be produced by the cells of a subject (e.g., a human), e.g., in the context of gene therapy, by administrating a vector (such as a viral vector (e.g., a retroviral vector, adenoviral vector, poxviral vector (e.g., vaccinia viral vector, such as Modified Vaccinia Ankara (MVA)), adeno-associated viral vector, and alphaviral vector)) containing a nucleic acid molecule encoding the polypeptide of the invention.
  • a vector such as a viral vector (e.g., a retroviral vector, adenoviral vector, poxviral vector (e.g., vaccinia viral vector, such as Modified Vaccinia Ankara (MVA)), adeno-associated viral vector, and alphaviral vector)
  • a vector such as a viral vector (e.g., a retroviral vector, aden
  • the vector once inside a cell of the subject (e.g., by transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, infection, etc.) will promote expression of the polypeptide, which is then secreted from the cell. If treatment of a disease or disorder is the desired outcome, no further action may be required. If collection of the protein is desired, blood may be collected from the subject and the protein purified from the blood by methods known in the art.
  • compositions that include the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)).
  • a pharmaceutical composition of the invention includes a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)) with a C-terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids) as the therapeutic protein.
  • a pharmaceutical composition of the invention includes a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety (e.g., a Fc domain monomer, or a dimer thereof, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin) as the therapeutic protein.
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • a moiety e.g., a Fc domain monomer, or a dimer thereof, a wild-type Fc domain,
  • a pharmaceutical composition of the invention including a polypeptide of the invention may be used in combination with other agents (e.g., therapeutic biologies and/or small molecules) or compositions in a therapy.
  • the pharmaceutical composition may include one or more pharmaceutically acceptable carriers or excipients, which can be formulated by methods known to those skilled in the art.
  • a pharmaceutical composition of the invention includes a nucleic acid molecule (DNA or RNA, e.g., mRNA) encoding a polypeptide of the invention, or a vector containing such a nucleic acid molecule.
  • Acceptable carriers and excipients in the pharmaceutical compositions are nontoxic to recipients at the dosages and concentrations employed.
  • Acceptable carriers and excipients may include buffers such as phosphate, citrate, HEPES, and TAE, antioxidants such as ascorbic acid and methionine, preservatives such as hexamethonium chloride, octadecyldimethylbenzyl ammonium chloride, resorcinol, and benzalkonium chloride, proteins such as human serum albumin, gelatin, dextran, and
  • compositions of the invention can be administered parenterally in the form of an injectable formulation.
  • Pharmaceutical compositions for injection can be formulated using a sterile solution or any pharmaceutically acceptable liquid as a vehicle.
  • Pharmaceutically acceptable vehicles include, but are not limited to, sterile water, physiological saline, and cell culture media (e.g., Dulbecco's Modified Eagle Medium (DMEM), a-Modified Eagles Medium (a-MEM), F-12 medium).
  • DMEM Dulbecco's Modified Eagle Medium
  • a-MEM a-Modified Eagles Medium
  • F-12 medium e.g., Banga (ed.) Therapeutic Peptides and Proteins: Formulation, Processing and Delivery Systems (3rd ed.) Taylor & Francis Group, CRC Press (2015).
  • the pharmaceutical compositions of the invention may be prepared in microcapsules, such as hydroxylmethylcellulose or gelatin-microcapsule and poly-(methylmethacrylate) microcapsule.
  • the pharmaceutical compositions of the invention may also be prepared in other drug delivery systems such as liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules. Such techniques are described in Remington: The Science and Practice of Pharmacy 22 th edition (2012).
  • the pharmaceutical compositions to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • compositions of the invention may also be prepared as a sustained-release formulation.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptides of the invention.
  • sustained release matrices include polyesters, hydrogels, polyactides, copolymers of L-glutamic acid and ⁇ ethyl-L- glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as LUPRON DEPOTTM, and poly-D-(-)-3-hydroxybutyric acid.
  • Some sustained-release formulations enable release of molecules over a few months, e.g., one to six months, while other formulations release pharmaceutical compositions of the invention for shorter time periods, e.g., days to weeks.
  • the pharmaceutical composition may be formed in a unit dose form as needed.
  • the amount of active component, e.g., a polypeptide of the invention, included in the pharmaceutical preparations is such that a suitable dose within the designated range is provided (e.g., a dose within the range of 0.01 - 100 mg/kg of body weight).
  • the pharmaceutical composition for gene therapy can be in an acceptable diluent, or can include a slow release matrix in which the gene delivery vehicle is imbedded. If hydrodynamic injection is used as the delivery method, the pharmaceutical composition containing a nucleic acid molecule encoding a polypeptide described herein or a vector (e.g., a viral vector) containing the nucleic acid molecule is delivered rapidly in a large fluid volume intravenously.
  • a vector e.g., a viral vector
  • Vectors that may be used as in vivo gene delivery vehicle include, but are not limited to, retroviral vectors, adenoviral vectors, poxviral vectors (e.g., vaccinia viral vectors, such as Modified Vaccinia Ankara), adeno-associated viral vectors, and alphaviral vectors.
  • retroviral vectors e.g., retroviral vectors, adenoviral vectors, poxviral vectors (e.g., vaccinia viral vectors, such as Modified Vaccinia Ankara), adeno-associated viral vectors, and alphaviral vectors.
  • compositions that include the polypeptides of the invention as the therapeutic proteins may be formulated for, e.g., intravenous administration, parenteral administration, subcutaneous administration, intramuscular administration, intra-arterial administration, intrathecal administration, or intraperitoneal administration.
  • the pharmaceutical composition may also be formulated for, or administered via, oral, nasal, spray, aerosol, rectal, or vaginal administration.
  • various effective pharmaceutical carriers are known in the art. See, e.g., ASHP Handbook on Injectable Drugs, Toissel, 18th ed. (2014).
  • a pharmaceutical composition that includes a nucleic acid molecule encoding a polypeptide of the invention or a vector containing such nucleic acid molecule may be administered by way of gene delivery.
  • Methods of gene delivery are well-known to one of skill in the art.
  • Vectors that may be used for in vivo gene delivery and expression include, but are not limited to, retroviral vectors, adenoviral vectors, poxviral vectors (e.g., vaccinia viral vectors, such as Modified Vaccinia Ankara (MVA)), adeno-associated viral vectors, and alphaviral vectors.
  • mRNA molecules encoding polypeptides of the invention may be administered directly to a subject.
  • nucleic acid molecules encoding a polypeptide described herein or vectors containing such nucleic acid molecules may be administered using a hydrodynamic injection platform.
  • a nucleic acid molecule encoding a polypeptide described herein is put under the control of a strong promoter in an engineered plasmid (e.g., a viral plasmid).
  • the plasmid is often delivered rapidly in a large fluid volume intravenously.
  • Hydrodynamic injection uses controlled hydrodynamic pressure in veins to enhance cell permeability such that the elevated pressure from the rapid injection of the large fluid volume results in fluid and plasmid extravasation from the vein.
  • the expression of the nucleic acid molecule is driven primarily by the liver.
  • hydrodynamic injection is often performed by injection of the plasmid into the tail vein.
  • mRNA molecules encoding a polypeptide described herein may be administered using hydrodynamic injection.
  • the dosage of the pharmaceutical compositions of the invention depends on factors including the route of administration, the disease to be treated, and physical characteristics, e.g., age, weight, general health, of the subject.
  • a pharmaceutical composition of the invention may include a dosage of a polypeptide of the invention ranging from 0.01 to 500 mg/kg (e.g., 0.01 , 0.1 , 0.2, 0.3, 0.4, 0.5, 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 1 50, 200, 250, 300, 350, 400, 450, or 500 mg/kg) and, in a more specific embodiment, about 0.1 to about 30 mg/kg and, in a more specific embodiment, about 0.3 to about 30 mg/kg.
  • the dosage may be adapted by the physician in accordance with conventional factors such as the extent of the disease and different parameters of the subject.
  • the pharmaceutical compositions are administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective to result in an improvement or remediation of the symptoms.
  • the pharmaceutical compositions are administered in a variety of dosage forms, e.g., intravenous dosage forms, subcutaneous dosage forms, and oral dosage forms (e.g., ingestible solutions, drug release capsules).
  • therapeutic proteins are dosed at 0.1 -100 mg/kg, e.g., 1 -50 mg/kg.
  • Pharmaceutical compositions that include a polypeptide of the invention may be administered to a subject in need thereof, for example, one or more times (e.g., 1 -10 times or more) daily, weekly, biweekly, monthly, bimonthly, quarterly, biannually, annually, or as medically necessary.
  • compositions that include a polypeptide of the invention may be administered to a subject in need thereof weekly, biweekly, monthly, bimonthly, or quarterly. Dosages may be provided in either a single or multiple dosage regimens. The timing between administrations may decrease as the medical condition improves or increase as the health of the patient declines.
  • the invention is based on the discovery that substituting amino acids from the extracellular portion of ActRllb into the extracellular portion ActRlla yields ActRlla variants with improved properties.
  • the ActRlla variants generated by introducing residues from ActRllb into ActRlla retain the beneficial properties of ActRlla, such as longer serum half-life and low binding affinity to BMP9, and gain some of the beneficial properties of ActRllb, such as increased binding to activins A and B (see Table 4).
  • These ActRlla variant properties produce a polypeptide that can be used therapeutically to compete with endogenous activin receptors for ligand binding.
  • the extracellular ActRlla variants contain the extracellular portion of the receptor, they are soluble and able to bind to and sequester ligands (e.g., activins A and B, myostatin, GDF1 1 ) without activating intracellular signaling pathways. Therefore, the extracellular ActRlla variants can be used to treat diseases or conditions in which elevated activin signaling has been implicated (e.g., associated with increased expression of activin receptors or activin receptor ligands). For example, activin has been found to be upregulated in bone disease and is known to inhibit osteoblast activity, suggesting that increased activin levels contribute to bone disease. It follows that treatment with a therapeutic agent that binds to activin and reduces its interaction with endogenous receptors could be used to increase bone mineral density and treat subjects with diseases or conditions involving bone damage.
  • sequester ligands e.g., activins A and B, myostatin, GDF1 1
  • the invention provides compositions and methods of treatment that may be used to increase bone mineral density, increase bone formation, or reduce bone resorption in a subject in need thereof.
  • the subject may have a disease that results in bone damage (e.g., osteoporosis or osteopenia).
  • the methods described herein are directed to affecting myostatin, activin, and/or BMP9 signaling in a subject having a disease or condition involving bone damage.
  • a polypeptide including an extracellular ActRlla variant described herein reduces or inhibits the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla).
  • affecting myostatin, activin, and/or BMP9 signaling results in an increase in the subject's bone mineral density or bone formation, or a decrease in the subject's bone resorption.
  • the polypeptides described herein may be administered to a subject to increase bone mineral density, to increase bone formation, to decrease bone resorption, or to affect myostatin, activin, and/or BMP9 signaling in the subject.
  • the methods described herein increase bone mineral density of the subject.
  • the methods described herein do not cause any vascular complications in the subject, such as increased vascular permeability or leakage.
  • the subject has a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss).
  • a disease or condition involving bone damage e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss).
  • bone damage e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metasta
  • the invention also includes methods of treating a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss by administering to the subject a polypeptide described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 - 72 (e.g., SEQ ID NOs: 6-72)).
  • a polypeptide described herein e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 - 72 (e.g., SEQ ID NOs:
  • the primary osteoporosis is age-related or hormone-related osteoporosis (e.g., related to a decline in estrogen).
  • the secondary osteoporosis is immobilization-induced or glucocorticoid-induced osteoporosis.
  • the bone cancer is multiple myeloma or the cancer metastasis-related bone loss is caused by multiple myeloma.
  • the treatment-related bone loss occurs due to treatment with FGF-21 or GLP-1 , treatment with an FGF-21 or GLP-1 containing therapeutic, or treatment of Type-2 diabetes and/or obesity, or due to cancer therapy (e.g., chemotherapy or radiation).
  • the diet-related bone loss is rickets (e.g., vitamin D deficiency).
  • the low-gravity related bone loss is lack of load-related bone loss.
  • the polypeptides described herein may be used to prevent the development of a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, osteopetrosis, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) and/or to treat patients already diagnosed with a disease or condition involving bone damage.
  • a disease or condition involving bone damage e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, osteopetrosis, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss
  • Patients who are likely to develop a disease or condition involving bone damage may be administered the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) prophylactically, such that the extracellular ActRlla polypeptides may prevent or delay the development of bone damage.
  • a polypeptide including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • the extracellular ActRlla polypeptides may prevent or delay the development of bone damage.
  • the polypeptides described herein may be administered to a subject to prevent the development of and/or treat patients with a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or to affect myostatin, activin, and/or BMP9 signaling in the subject (e.g., to reduce or inhibit the binding of activin, myostatin, and/or BMP9 to their receptors
  • a disease or condition involving bone damage e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-
  • the methods described herein increase bone mineral density (e.g., increase bone mass). In some embodiments, the methods described herein reduce bone resorption (e.g., reduce bone catabolic activity). In some embodiments, the methods described herein increase bone formation (e.g., increase bone anabolic activity or increase osteogenesis). In some embodiments, the methods described herein increase osteoblast activity or osteoblastogenesis. In some embodiments, the methods described herein decrease osteoclast activity or osteoclastogenesis. In some embodiments, the methods described herein reduce or inhibit the binding of activin and/or myostatin to their receptors. In some embodiments, the methods increase bone formation, increase bone mineral density, or decrease bone resorption of cortical or trabecular bone.
  • a polypeptide including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -71 (e.g., SEQ ID NOs: 6-71 )
  • an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -71 (e.g., SEQ ID NOs: 6-71 )
  • a C-terminal extension of one or more amino acids e.g., 1 , 2, 3, 4, 5, 6, or more amino acids
  • a dimer e.g., homodimer or heterodimer
  • a polypeptide including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)
  • an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72) fused to an Fc domain monomer
  • a polypeptide including an extracellular ActRlla variant e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety (e.g., a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin) may be used as the therapeutic protein.
  • Nucleic acids encoding the polypeptides described herein, or vectors containing said nucleic acids can also be administered according to any of the methods described herein.
  • the polypeptide, nucleic acid, or vector can be administered as part of a pharmaceutical composition.
  • the Biacore 3000 was used to measure the kinetics of the interactions between the ActRlla variants and the ligands Activin A, Activin B, growth differentiation factor 1 1 (GDF1 1 ), and BMP-9.
  • ActRlla variants were transiently expressed in HEK293 cells and purified from the conditioned media using Protein-A Sepharose chromatography.
  • the ActRlla variants were immobilized on the chip (CM4 or CM5) with capture antibodies (anti-mouse from GEGE) in flow cells 2-4 to ensure proper orientation.
  • Flow cell 1 was used as a reference cell to subtract any nonspecific binding and bulk effects.
  • HBS-EP+ buffer from GE HealthcareTM was used as a running buffer. Each ligand was run in a concentration series at 40 ⁇ /min to avoid mass transport effects. The data was analyzed using Scrubber2 by BioLogicTM Software to calculate the KD of each interaction (Table 4).
  • tissues of interest (muscles, fat depots, and tibias) are surgically removed, weighed, and properly stored for further analysis.
  • the ORX animals are also examined to confirm complete removal of testes.
  • Cortical morphometry and trabecular structure of the various bones are also evaluated after the experiment termination using micro-computed tomography.

Abstract

The invention features polypeptides that include an extracellular ActRlla variant. In some embodiments, a polypeptide of the invention includes an extracellular ActRlla variant fused to an Fc domain monomer or moiety. The invention also features pharmaceutical compositions and methods of using the polypeptides to treat diseases and conditions involving bone damage, e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.

Description

ACTIVIN RECEPTOR TYPE MA VARIANTS AND METHODS OF USE THEREOF
BACKGROUND OF THE INVENTION
Healthy bone undergoes a constant remodeling that involves both bone breakdown and bone growth. Bone growth is mediated by the osteoblast cell type whereas the osteoclasts resorb the bone Pathology occurs when these systems fall out of balance either through downregulation of the anabolic program, upregulation of the catabolic system or a combination of both, resulting in a net bone loss. Therefore, controlling the balance in bone remodeling can be useful for promoting the healing of fractures and other damage to bone as well as the treatment of disorders, such as osteoporosis, associated with loss of bone mass and bone mineralization.
Bone damage can result from a range of root causes, including age- or cancer-related bone loss, genetic conditions, adverse side effects of drug treatment, or fracture. The World Health Organization estimates that osteoporosis alone affects 75 million people in the U.S., Europe and Japan, and is a significant risk factor in bone fracture. In general, the whole of bone loss represents pathological states for which there are few effective treatments. Treatment instead focuses on immobilization, exercise and dietary modifications rather than agents that directly promote bone growth and increase bone density. With respect to osteoporosis, estrogen, calcitonin, osteocalcin with vitamin K, or high doses of dietary calcium are all used as therapeutic interventions. Other therapeutic approaches to osteoporosis include bisphosphonates, parathyroid hormone, parathyroid hormone related protein (PTHrP) calcimimetics, statins, anabolic steroids, lanthanum and strontium salts, and sodium fluoride. Such therapeutics, however, are often associated with undesirable side effects. There exists a need for novel and effective treatments for diseases that result in bone damage or bone demineralization.
SUMMARY OF THE INVENTION
The present invention features polypeptides that include an extracellular activin receptor type lla
(ActRlla) variant. In some embodiments, a polypeptide of the invention includes an extracellular ActRlla variant fused to the N- or C-terminus of an Fc domain monomer or moiety. Such moieties may be attached by amino acid or other covalent bonds. A polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may also form a dimer (e.g., a homodimer or heterodimer) through the interaction between two Fc domain monomers. The polypeptides of the invention may be used to increase bone mass or bone mineral density in a subject having a disease or condition involving bone damage, e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility.. Further, the polypeptides of the invention may also be used to affect myostatin, activin, and/or bone morphogenetic protein 9 (BMP9) signaling in a subject having a risk of developing or having a disease or condition involving bone damage or bone demineralization.
In one aspect, the invention features a polypeptide including an extracellular activin receptor type lla (ActRlla) variant, the variant having a sequence of
GAILG RSETQECLX1X2NANWX3X4X5X6TNQTGVEX7CX8GX9X10X11X12X13X14HCX15ATWX16NISGSI EIVX1 7X18GCX19X20X21 DX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCN EKFSYFPEMEVTQPTS (SEQ ID NO: 1 ), wherein Xi is F or Y; X2 is F or Y; X3 is E or A; X4 is K or L; X5 is D or E ; X6 is R or A; X7 is P or R; Xs is Y or E; X9 is D or E ; X10 is K or Q; Xn is D or A; X12 is K or A; X13 is R or A; X14 is R or L; X15 is F or Y; X16 is K, R, or A; X17 is K, A, Y, F, or I ; Xi8 is Q or K; X19 is W or A; X20 is L or A; X21 is D, K, R, A, F, G, M, N, or I; X22 is I, F, or A; X23 is K or T; X24 is K or E; X25 is D or E ; X26 is S or N; and X27 is E or Q, and wherein the variant has at least one amino acid substitution relative to a wild-type extracellular ActRlla having the sequence of SEQ ID NO: 73 or an extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96.
In some embodiments, the variant has a sequence of
GAILG RSETQECLFX2NANWX3X4X5X6TNQTGVEX7CX8GX9KX11 X12X13X14HCX15ATWX16NISGSIEIVX17X1 8GCX19X20X21 DX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 2), wherein X2, X3, X4, Xs, Χβ, X7, Xs, X9, Xn , X12, X13, X14, X15, X16, X17, Xis, X19, X20, X21 , X22, X23, X24, X25, X26, and X27 are defined as above.
In some embodiments, the variant has a sequence of
GAILG RSETQECLFX2NANWEX4X5RTNQTGVEX7CX8GX9KDKRX14HCX15ATWX16NISGSI EIVKX18GCWL DDX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 3), wherein X2, X4, Xs, X7, Xs, X9, X14, X15, X16, Xis, X22, X23, X24, X25, X26, and X27 are defined as above.
In some embodiments, the variant has a sequence of
GAILG RSETQECLFX2NANWEX4DRTNQTGVEX7CXsGX9KDKRXi4HCXi5ATWX16NISGSIEIVKXi8GCWL DDX22NCYDRTDCVEX23KX25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 4), wherein X2, X4, X7, Xs, X9, X14, X15, X16, Xis, X22, X23, X25, X26, and X27 are defined as above.
In some embodiments, the variant has a sequence of
GAILG RSETQECLFX2NANWEX4DRTNQTGVEPCX8GX9KDKRX14HCFATWKNISGSIEIVKX18GCWLDDI NCYDRTDCVEX23KX25X26PX27VYFCCCEGNMCN EKFSYFPEMEVTQPTS (SEQ ID NO: 5), wherein X2, X4, Xs, X9, Xi4, Xis, X23, X25, X26, and X27 are defined as above.
In any of the aforementioned embodiments, Xi is F or Y. In any of the aforementioned embodiments, X2 is F or Y. In any of the aforementioned embodiments, X3 is E or A. In any of the aforementioned embodiments, X4 is K or L. In any of the aforementioned embodiments, X5 is D or E. In any of the aforementioned embodiments, Χβ is R or A. In any of the aforementioned embodiments, X7 is P or R. In any of the aforementioned embodiments, Xs is Y or E. In any of the aforementioned embodiments, X9 is D or E. In any of the aforementioned embodiments, X10 is K or Q. In any of the aforementioned embodiments, Xn is D or A. In any of the aforementioned embodiments, X12 is K or A. In any of the aforementioned embodiments, X13 is R or A. In any of the aforementioned embodiments, Xi4 is R or L. In any of the aforementioned embodiments, X15 is F or Y. In any of the aforementioned embodiments, X16 is K, R, or A. In any of the aforementioned embodiments, X17 is K, A, Y, F, or I. In any of the aforementioned embodiments, Xis is Q or K. In any of the aforementioned embodiments, X19 is W or A. In any of the aforementioned embodiments, X20 is L or A. In any of the aforementioned
embodiments, X21 is D, K, R, A, F, G, M, N, or I. In any of the aforementioned embodiments, X22 is I, F, or A. In any of the aforementioned embodiments, X23 is K or T. In any of the aforementioned embodiments, X24 is K or E. In any of the aforementioned embodiments, X25 is D or E. In any of the aforementioned embodiments, X26 is S or N. In any of the aforementioned embodiments, X27 is E or Q. In any of the aforementioned embodiments, X23 is T, X24 is E, X25 is E, and X26 is N or X23 is T, X24 is K, X25 is E, and X26 is N. In any of the aforementioned embodiments, X17 is K.
In any of the aforementioned embodiments, the variant has the sequence of any one of SEQ ID NOs: 6-72.
In any of the aforementioned embodiments, the amino acid at position X24 may be replaced with the amino acid K.
In any of the aforementioned embodiments, the amino acid at position X24 may be replaced with the amino acid E.
In any of the aforementioned embodiments, a polypeptide described herein may further include a C-terminal extension of one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6 more amino acids). In some embodiments, the C-terminal extension is amino acid sequence NP. In some embodiments, the C- terminal extension is amino acid sequence NPVTPK (SEQ ID NO: 155).
In any of the aforementioned embodiments, a polypeptide described herein may further include a moiety fused or covalently linked to the C-terminus of the polypeptide. The moiety may increase the stability of improve pharmacokinetic properties of the polypeptide. In some embodiments, the moiety is an Fc domain monomer, an Fc domain, an albumin binding peptide, a fibronectin domain, or serum albumin.
In any of the aforementioned embodiments, a polypeptide described herein may further include an Fc domain monomer fused to the C-terminus of the polypeptide by way of a linker. In some embodiments, the polypeptide that includes an extracellular ActRMa variant described herein fused to an Fc domain monomer may form a dimer (e.g., a homodimer or heterodimer) through the interaction between two Fc domain monomers. In some embodiments, the Fc domain monomer has the sequence of SEQ ID NO: 97
In any of the aforementioned embodiments, a polypeptide described herein may further include an Fc domain fused to the C-terminus of the polypeptide by way of a linker. In some embodiments, the Fc domain is a wild-type Fc domain. In some embodiments, the wild-type Fc domain has the sequence of SEQ ID NO: 151 . In some embodiments, the Fc domain contains one or more amino acid substitutions. In some embodiments, the Fc domain containing one or more amino acid substitutions does not form a dimer.
In any of the aforementioned embodiments, a polypeptide described herein may further include an albumin-binding peptide fused to the C-terminus of the polypeptide by way of a linker. In some embodiments, the albumin-binding peptide has the sequence of SEQ ID NO: 1 52.
In any of the aforementioned embodiments, a polypeptide described herein may further include a fibronectin domain fused to the C-terminus of the polypeptide by way of a linker. In some embodiments, the fibronectin domain peptide has the sequence of SEQ ID NO: 153.
In any of the aforementioned embodiments, a polypeptide described herein may further include a human serum albumin fused to the C-terminus of the polypeptide by way of a linker. In some
embodiments, the human serum albumin has the sequence of SEQ ID NO: 154.
In some embodiments, the linker is an amino acid spacer. In some embodiments, the amino acid spacer is GGG, GGGA (SEQ ID NO: 98), GGGG (SEQ ID NO: 1 00), GGGAG (SEQ ID NO: 130), GGGAGG (SEQ ID NO: 131 ), or GGGAGGG (SEQ ID NO: 132). In some embodiments, the amino acid spacer is GGGS (SEQ ID NO: 99), GGGGA (SEQ ID NO: 101 ), GGGGS (SEQ ID NO: 102), GGGGG (SEQ ID NO: 103), GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), AGGG (SEQ ID NO: 106), SGGG (SEQ ID NO: 107), GAGA (SEQ ID NO: 108), GSGS (SEQ ID NO: 109), GAGAGA (SEQ ID NO: 1 10), GSGSGS (SEQ ID NO: 1 1 1 ), GAGAGAGA (SEQ ID NO: 1 12), GSGSGSGS (SEQ ID NO: 1 13), GAGAGAGAGA (SEQ ID NO: 1 14), GSGSGSGSGS (SEQ ID NO: 1 15), GAGAGAGAGAGA (SEQ ID NO: 1 16), and GSGSGSGSGSGS (SEQ ID NO: 1 1 7), GGAGGA (SEQ ID NO: 1 18), GGSGGS (SEQ ID NO: 1 19), GGAGGAGGA (SEQ ID NO: 120), GGSGGSGGS (SEQ ID NO: 121 ), GGAGGAGGAGGA (SEQ ID NO: 122), GGSGGSGGSGGS (SEQ ID NO: 123), GGAGGGAG (SEQ ID NO: 124), GGSGGGSG (SEQ ID NO: 125), GGAGGGAGGGAG (SEQ ID NO: 126), and GGSGGGSGGGSG (SEQ ID NO: 127), GGGGAGGGGAGGGGA (SEQ ID NO: 128),
GGGGSGGGGSGGGGS (SEQ ID NO: 129), AAAL (SEQ ID NO: 133), AAAK (SEQ ID NO: 134), AAAR (SEQ ID NO: 135), EGKSSGSGSESKST (SEQ ID NO: 136), G SAG S A AG SG E F (SEQ ID NO: 137), AEAAAKEAAAKA (SEQ ID NO: 138), KESGSVSSEQLAQFRSLD (SEQ ID NO: 139), GENLYFQSGG (SEQ ID NO: 140), SACYCELS (SEQ ID NO: 141 ), RSIAT (SEQ ID NO: 142), RPACKIPNDLKQKVMNH (SEQ ID NO: 143), GGSAGGSGSGSSGGSSGASGTGTAGGTGSGSGTGSG (SEQ ID NO: 144),
AAANSSIDLISVPVDSR (SEQ ID NO: 145), GGSGGGSEGGGSEGGGSEGGGSEGGGSEGGGSGGGS (SEQ ID NO: 146), EAAAK (SEQ ID NO: 147), or PAPAP(SEQ ID NO: 148).
In any of the aforementioned embodiments, the polypeptide described herein has a serum half- life of at least 7 days.
In any of the aforementioned embodiments, the polypeptide described herein binds to human bone morphogenetic protein 9 (BMP9) with a KD of 200 pM or higher. In some embodiments, the polypeptide binds to activin and/or myostatin and has reduced (e.g., weak) binding to human BMP9. In some embodiments, the polypeptide does not substantially bind to human BMP9.
In any of the aforementioned embodiments, the polypeptide described herein binds to human activin A with a KD of 800 pM or less.
In any of the aforementioned embodiments, the polypeptide described herein binds to human activin B with a KD of approximately 800 pM or less.
In any of the aforementioned embodiments, the polypeptide described herein binds to human GDF-1 1 with a KD of approximately 5 pM or higher.
In another aspect, the invention features a nucleic acid molecule encoding a polypeptide described herein (e.g., a polypeptide including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)). In another aspect, the invention also features a vector including the nucleic acid molecule described herein.
In another aspect, the invention features a host cell that expresses a polypeptide described herein, wherein the host cell includes a nucleic acid molecule or a vector described in the previous two aspects, wherein the nucleic acid molecule or vector is expressed in the host cell.
In another aspect, the invention features a method of preparing a polypeptide described herein, wherein the method includes: a) providing a host cell including a nucleic acid molecule or a vector described herein, and b) expressing the nucleic acid molecule or vector in the host cell under conditions that allow for the formation of the polypeptide. In another aspect, the invention features a pharmaceutical composition including a polypeptide, nucleic acid molecule, or vector described herein and one or more pharmaceutically acceptable carriers or excipients. In some embodiments of the pharmaceutical composition, the polypeptide, nucleic acid molecule, or vector is in a therapeutically effective amount.
In another aspect, the invention also features a construct including two identical polypeptides
(e.g., a homodimer) each including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to the N- or C-terminus of an Fc domain monomer (e.g., the sequence of SEQ ID NO: 97). The two Fc domain monomers in the two polypeptides interact to form an Fc domain in the construct.
In another aspect, the invention also features a construct including two different polypeptides
(e.g., a heterodimer) each including an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to the N- or C-terminus of an Fc domain monomer. The two Fc domain monomers in the two polypeptides interact to form an Fc domain in the construct.
In another aspect, the invention features a method of increasing bone mineral density in a subject in need thereof. The method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of reducing bone resorption in a subject in need thereof. The method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of increasing bone formation in a subject in need thereof. The method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In some embodiments of any of the above aspects, the subject has primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.
In another aspect, the invention features a method of affecting myostatin, activin, and/or BMP9 signaling (e.g., reducing or inhibiting the binding of myostatin, activin, and/or BMP9 to their receptors) in a subject having a disease or condition involving bone damage, wherein method includes administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein. In some embodiments of this aspect, the disease or condition is primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment- related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity- related bone loss, or immobility-related bone loss. In another aspect, the invention features a method of treating a subject having primary osteoporosis by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having secondary osteoporosis by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having osteopenia by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having a fracture by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having bone cancer or cancer metastasis-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having Paget's disease by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject renal osteodystrophy by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having treatment-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having diet-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having bone loss associated with the treatment of obesity by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having low gravity-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In another aspect, the invention features a method of treating a subject having immobility-related bone loss by administering to the subject a therapeutically effective amount of a polypeptide, nucleic acid molecule, or vector described herein or a pharmaceutical composition described herein.
In some embodiments of any of the above aspects, the primary osteoporosis is age-related osteoporosis. In some embodiments of any of the above aspects, the primary osteoporosis is hormone-related osteoporosis.
In some embodiments of any of the above aspects, the secondary osteoporosis is immobilization- induced osteoporosis.
In some embodiments of any of the above aspects, wherein the secondary osteoporosis is glucocorticoid-induced osteoporosis.
In some embodiments of any of the above aspects, the cancer is multiple myeloma.
In some embodiments of any of the above aspects, the treatment is FGF-21 treatment.
In some embodiments of any of the above aspects, the treatment is GLP-1 treatment.
In some embodiments of any of the above aspects, the treatment is cancer therapy.
In some embodiments of any of the above aspects, the treatment is treatment for obesity and/or Type-2 diabetes.
In some embodiments of any of the above aspects, the diet-related bone loss is rickets.
In some embodiments of any of the above aspects, the method increases bone formation in the subject. In some embodiments of any of the above aspects, wherein the method decreases bone resorption in the subject. In some embodiments of any of the above aspects, the method increases osteoblast activity or osteoblastogenesis. In some embodiments of any of the above aspects, the method decreases osteoclast activity or decreases osteoclastogenesis. In some embodiments of any of the above aspects, the method reduces or inhibits the binding of activin and/or myostatin to their receptors.
In some embodiments of any of the methods described herein, the method does not cause a vascular complication (e.g., an increase vascular permeability or leakage) in the subject. In some embodiments of any of the methods described herein, the method increases bone mineral density in the subject.
In some embodiments of any of the above aspects, the bone is cortical bone. In some embodiments of any of the above aspects, the bone is trabecular bone.
In some embodiments of any of the above aspects, the polypeptide, nucleic acid, vector, or pharmaceutical composition is administered in an amount sufficient to increase bone density, reduce bone resorption, reduce the rate of bone resorption, increase bone formation, increase the rate of bone formation, reduce osteoclast activity, increase osteoblast activity, or affect myostatin, activin, and/or BMP9 signaling in the subject.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 69. In some embodiments, the variant having the sequence of SEQ ID NO: 69 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 69, optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 58. In some embodiments, the variant having the sequence of SEQ ID NO: 58 has the amino acid K at position X17, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 58, optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 6. In some embodiments, the variant having the sequence of SEQ ID NO: 6 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 6, optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C-terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 38. In some embodiments, the variant having the sequence of SEQ ID NO: 38 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155 In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 38, optionally having the amino acid K at position X17, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C-terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 41 . In some embodiments, the variant having the sequence of SEQ ID NO: 41 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 41 , optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 44. In some embodiments, the variant having the sequence of SEQ ID NO: 44 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 44, optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 70. In some embodiments, the variant having the sequence of SEQ ID NO: 70 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids NP or NPVTPK (SEQ ID NO: 155)). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 70, optionally having the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 71 . In some embodiments, the variant having the sequence of SEQ ID NO: 71 has the amino acid K at position Xi7, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C- terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids at the C-terminus, e.g., the amino acids VTPK). In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 71 , optionally having the amino acid K at position X17, the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26, and/or a C-terminal extension, or a pharmaceutical composition containing said variant.
In some embodiments of any of the above aspects, the variant has the sequence of SEQ ID NO: 72. In some embodiments, the variant having the sequence of SEQ ID NO: 72 has the amino acid K at position Xi7 and/or the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26. In some embodiments of any of the above aspects, the method includes increasing bone mineral density, increasing bone formation, decreasing bone resorption, or treating a condition or disease involving bone damage in a subject in need thereof (e.g., a subject having primary osteoporosis, secondary
osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or affecting myostatin, activin, and/or BMP9 signaling in a subject (e.g., a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) by administering to the subject a therapeutically effective amount of a variant having the sequence of SEQ ID NO: 72, optionally having the amino acid K at position X17 and/or the amino acid sequence TEEN or TKEN at positions X23, X24, X25, and X26.
Definitions
As used herein, the term "extracellular activin receptor type Ma (ActRlla) variant" refers to a peptide including a soluble, extracellular portion of the single transmembrane receptor, ActRlla, that has at least one amino acid substitution relative to a wild-type extracellular ActRlla (e.g., bold portion of the sequence of SEQ ID NO: 75 shown below) or an extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96. The sequence of the wild-type, human ActRlla precursor protein is shown below (SEQ ID NO: 75), in which the signal peptide is italicized and the extracellular portion is bold. Wild-type, human ActRlla precursor protein (SEQ ID NO: 75):
i FZ./SCSSGAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFAT WKNISGSIEIVKQGCWLDDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPT
SNPVTPKPPYYNILLYSLVPLMLIAGIVICAFWVYRHHKMAYPPVLVPTQDPGPPPPSPLLGLKPL QLLEVKARGRFGCVWKAQLLNEYVAVKIFPIQDKQSWQNEYEVYSLPGMKHENILQFIGAEKRG TSVDVDLWLITAFHEKGSLSDFLKANVVSWNELCHIAETMARGLAYLHEDI PGLKDGHKPAISHR DIKSKNVLLKNNLTACIADFGLALKFEAGKSAGDTHGQVGTRRYMAPEVLEGAINFQRDAFLRID MYAMGLVLWELASRCTAADGPVDEYMLPFEEEIGQHPSLEDMQEVVVHKKKRPVLRDYWQKH AGMAMLCETIEECWDHDAEARLSAGCVGERITQMQRLTNIITTEDIVTVVTMVTNVDFPPKESSL
An extracellular ActRlla variant may have a sequence of any one of SEQ ID NOs: 1 -72. In particular embodiments, an extracellular ActRlla variant has a sequence of any one of SEQ ID NOs: 6-72 (Table 2). In some embodiments, an extracellular ActRlla variant may have at least 85% (e.g., at least 85%, 87%, 90%, 92%, 95%, 97%, or greater) amino acid sequence identity to the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73).
As used herein, the term "extracellular ActRllb variant" refers to a peptide including a soluble, extracellular portion of the single transmembrane receptor, ActRllb, that has at least one amino acid substitution relative to a wild-type extracellular ActRllb (e.g., the sequence of SEQ ID NO: 74). An extracellular ActRllb variant may have the sequence of SEQ ID NO: 149 shown below:
extracellular ActRllb variant (SEQ ID NO: 149):
GRGEAETRECIFYNANWEKDRTNQSGLEPCYGDQDKRRHCFASWKNSSGTIELVKQGCWLDDI
NCYDRQECVAKKDSPEVYFCCCEGNFCNERFTHLPEAGGPEVTYEPPPTAPT
As used herein, the term "linker" refers to a linkage between two elements, e.g., peptides or protein domains. A polypeptide described herein may include an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)) fused to a moiety. The moiety may increase stability or improve pharmacokinetic properties of the polypeptide. The moiety (e.g., Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin) may be fused to the polypeptide by way of a linker. A linker can be a covalent bond or a spacer. The term "bond" refers to a chemical bond, e.g., an amide bond or a disulfide bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation. The term "spacer" refers to a moiety (e.g., a polyethylene glycol (PEG) polymer) or an amino acid sequence (e.g., a 1 -200 amino acid sequence) occurring between two elements, e.g., peptides or protein domains, to provide space and/or flexibility between the two elements. An amino acid spacer is part of the primary sequence of a polypeptide (e.g., fused to the spaced peptides via the polypeptide backbone). The formation of disulfide bonds, e.g., between two hinge regions that form an Fc domain, is not considered a linker.
As used herein, the term "Fc domain" refers to a dimer of two Fc domain monomers. An Fc domain has at least 80% sequence identity (e.g., at least 85%, 90%, 95%, 97%, or 100% sequence identity) to a human Fc domain that includes at least a CH2 domain and a CH3 domain. An Fc domain monomer includes second and third antibody constant domains (CH2 and CH3). In some embodiments, the Fc domain monomer also includes a hinge domain. An Fc domain does not include any portion of an immunoglobulin that is capable of acting as an antigen-recognition region, e.g., a variable domain or a complementarity determining region (CDR). In the wild-type Fc domain, the two Fc domain monomers dimerize by the interaction between the two CH3 antibody constant domains, as well as one or more disulfide bonds that form between the hinge domains of the two dimerizing Fc domain monomers. In some embodiments, an Fc domain may be mutated to lack effector functions, typical of a "dead Fc domain." In certain embodiments, each of the Fc domain monomers in an Fc domain includes amino acid substitutions in the CH2 antibody constant domain to reduce the interaction or binding between the Fc domain and an Fey receptor. In some embodiments, the Fc domain contains one or more amino acid substitutions that reduce or inhibit Fc domain dimerization. An Fc domain can be any immunoglobulin antibody isotype, including IgG, IgE, IgM, IgA, or IgD. Additionally, an Fc domain can be an IgG subtype (e.g., lgG1 , lgG2a, lgG2b, lgG3, or lgG4). The Fc domain can also be a non-naturally occurring Fc domain, e.g., a recombinant Fc domain.
As used herein, the term "albumin-binding peptide" refers to an amino acid sequence of 12 to 16 amino acids that has affinity for and functions to bind serum albumin. An albumin-binding peptide can be of different origins, e.g., human, mouse, or rat. In some embodiments, an albumin-binding peptide has the sequence DICLPRWGCLW (SEQ ID NO: 1 52).
As used herein, the term "fibronectin domain" refers to a high molecular weight glycoprotein of the extracellular matrix, or a fragment thereof, that binds to, e.g., membrane-spanning receptor proteins such as integrins and extracellular matrix components such as collagens and fibrins. In some embodiments, a fibronectin domain is a fibronectin type III domain (SEQ ID NO: 153) having amino acids 61 0-702 of the sequence of UniProt ID NO: P02751 . In other embodiments, a fibronectin domain is an adnectin protein.
As used herein, the term "human serum albumin" refers to the albumin protein present in human blood plasma. Human serum albumin is the most abundant protein in the blood. It constitutes about half of the blood serum protein. In some embodiments, a human serum albumin has the sequence of UniProt ID NO: P02768 (SEQ ID NO: 154).
As used herein, the term "fused" is used to describe the combination or attachment of two or more elements, components, or protein domains, e.g., peptides or polypeptides, by means including chemical conjugation, recombinant means, and chemical bonds, e.g., amide bonds. For example, two single peptides in tandem series can be fused to form one contiguous protein structure, e.g., a polypeptide, through chemical conjugation, a chemical bond, a peptide linker, or any other means of covalent linkage. In some embodiments of a polypeptide described herein, an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused in tandem series to the N- or C-terminus of a moiety (e.g., Fc domain monomer (e.g., the sequence of SEQ ID NO: 97) a wild-type Fc domain (e.g., the sequence of SEQ ID NO: 151 ), an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide (e.g., the sequence of SEQ ID NO: 152), a fibronectin domain (e.g., the sequence of SEQ ID NO: 1 53), or a human serum albumin (e.g., the sequence of SEQ ID NO: 154)) by way of a linker. For example, an extracellular ActRlla variant is fused to a moiety (e.g., an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin) by way of a peptide linker, in which the N-terminus of the peptide linker is fused to the C- terminus of the extracellular ActRlla variant through a chemical bond, e.g., a peptide bond, and the C- terminus of the peptide linker is fused to the N-terminus of the moiety (e.g., Fc domain monomer, wild- type Fc domain, Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), albumin-binding peptide, fibronectin domain, or human serum albumin) through a chemical bond, e.g., a peptide bond.
As used herein, the term "C-terminal extension" refers to the addition of one or more amino acids to the C-terminus of a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)). The C-terminal extension can be one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, or more amino acids). Exemplary C- terminal extensions are the amino acid sequence NP (a two amino acid C-terminal extension) and the amino acid sequence NPVTPK (SEQ ID NO: 1 55) (a six amino acid C-terminal extension). Any amino acid sequence that does not disrupt the activity of the polypeptide can be used. SEQ ID NO: 71 , which is the sequence of SEQ ID NO: 69 with a C-terminal extension of NP, and SEQ ID NO: 72, which is the sequence of SEQ ID NO: 69 with a C-terminal extension of NPVTPK, represent two of the possible ways that a polypeptide of the invention can be modified to include a C-terminal extension.
As used herein, the term "percent (%) identity" refers to the percentage of amino acid (or nucleic acid) residues of a candidate sequence, e.g., an extracellular ActRlla variant, that are identical to the amino acid (or nucleic acid) residues of a reference sequence, e.g., a wild-type extracellular ActRlla (e.g., SEQ ID NO: 73), after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity (i.e., gaps can be introduced in one or both of the candidate and reference sequences for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). Alignment for purposes of determining percent identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, ALIGN, or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. In some embodiments, the percent amino acid (or nucleic acid) sequence identity of a given candidate sequence to, with, or against a given reference sequence (which can alternatively be phrased as a given candidate sequence that has or includes a certain percent amino acid (or nucleic acid) sequence identity to, with, or against a given reference sequence) is calculated as follows:
100 x (fraction of A/B)
where A is the number of amino acid (or nucleic acid) residues scored as identical in the alignment of the candidate sequence and the reference sequence, and where B is the total number of amino acid (or nucleic acid) residues in the reference sequence. In some embodiments where the length of the candidate sequence does not equal to the length of the reference sequence, the percent amino acid (or nucleic acid) sequence identity of the candidate sequence to the reference sequence would not equal to the percent amino acid (or nucleic acid) sequence identity of the reference sequence to the candidate sequence. In particular embodiments, a reference sequence aligned for comparison with a candidate sequence may show that the candidate sequence exhibits from 50% to 100% identity across the full length of the candidate sequence or a selected portion of contiguous amino acid (or nucleic acid) residues of the candidate sequence. The length of the candidate sequence aligned for comparison purpose is at least 30%, e.g., at least 40%, e.g., at least 50%, 60%, 70%, 80%, 90%, or 100% of the length of the reference sequence. When a position in the candidate sequence is occupied by the same amino acid (or nucleic acid) residue as the corresponding position in the reference sequence, then the molecules are identical at that position.
As used herein, the term "serum half-life" refers to, in the context of administering a therapeutic protein to a subject, the time required for plasma concentration of the protein in the subject to be reduced by half. The protein can be redistributed or cleared from the bloodstream, or degraded, e.g., by proteolysis. As described herein, a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)) displays a serum half-life of 7 days in humans. As used herein, the term "affinity" or "binding affinity" refers to the strength of the binding interaction between two molecules. Generally, binding affinity refers to the strength of the sum total of non-covalent interactions between a molecule and its binding partner, such as an extracellular ActRlla variant and BMP9 or activin A. Unless indicated otherwise, binding affinity refers to intrinsic binding affinity, which reflects a 1 :1 interaction between members of a binding pair. The binding affinity between two molecules is commonly described by the dissociation constant (KD) or the affinity constant (KA). TWO molecules that have low binding affinity for each other generally bind slowly, tend to dissociate easily, and exhibit a large KD. TWO molecules that have high affinity for each other generally bind readily, tend to remain bound longer, and exhibit a small KD. The KD of two interacting molecules may be determined using methods and techniques well known in the art, e.g., surface plasmon resonance. KD is calculated as the ratio of k0ff/kon.
As used herein, the terms "bone mineral density (BMD)" "bone density" and "bone mass" refer to a measure of the amount of bone mineral (e.g. calcium) in bone tissue. BMD may be measured by well- established clinical techniques known to one of skill in the art (e.g., by single-1 or dual-energy photon or X-ray absorptiometry). The concept of BMD relates to the mass of mineral per volume of bone, although clinically it is measured by proxy according to optical density per square centimeter of bone surface upon imaging. BMD measurement is used in clinical medicine as an indirect indicator of osteoporosis and fracture risk. In some embodiments, BMD test results are provided as a T-score, where the T-score represents the BMD of a subject compared to the ideal or peak bone mineral density of a healthy 30-year- old adult. A score of 0 indicates that the BMD is equal to the normal reference value for a healthy young adult. Differences between the measured BMD of subject and that of the reference value for a healthy young adult are measured in standard deviations units (SDs). Accordingly, a T-score of between +1 SD and -1 SD may indicate a normal BMD, a T-score of between -1 SD and -2.5 SD may indicate low bone mass (e.g., osteopenia), and a T-score lower than -2.5 SD may indicate osteoporosis or severe osteoporosis. In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, wherein the patient has low bone mass (e.g., a T-Score of between -1 SD and -2.5 SD). In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, wherein the patient has osteoporosis (e.g., a T-Score of less than -2.5 SD). In some embodiments, administration of a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule treats the subject by increasing their BMD. In some embodiments, administration of a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule increases the BMD of a subject resulting in an increase in the T- Score of the subject (e.g., resulting in an increase in the T-Score of the subject of 0.1 or more, 0.2 or more, 0.3 or more, 0.4 or more, 0.5 or more, 1 .0 or more, or 2.0 or more).
As used herein, the terms "bone remodeling" or "bone metabolism" refer to the process for maintaining bone strength and ion homeostasis by replacing discrete parts of old bone with newly synthesized packets of proteinaceous matrix. Bone is resorbed by osteoclasts, and is deposited by osteoblasts in a process called ossification. Osteocyte activity plays a key role in this process.
Conditions that result in a decrease in bone mass, can either be caused by an increase in resorption, or a decrease in ossification. In a healthy individual, during childhood, bone formation exceeds resorption. As the aging process occurs, resorption exceeds formation. Bone resorption rates are also typically much higher in post-menopausal older women due to estrogen deficiency related to menopause.
As used herein, the terms "bone resorption" or "bone catabolic activity" refer to a process by which osteoclasts break down the tissue in bones and release the minerals, resulting in a transfer of the mineral (e.g., calcium) from bone tissue to the blood. Increased rates of bone resorption are associated with aging, including in post-menopausal women. High rates of bone resorption, or rates of bone resorption that exceed the rate of ossification, are associated with bone disorders, such as decreased bone mineral density, including osteopenia and osteoporosis. In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof to decrease done resorption in the subject (e.g., the rate of bone resorption in the subject).
As used herein, the terms "bone formation," "ossification," "osteogenesis," or "bone anabolic activity" refer to the process of forming new bone tissue by osteoblasts. In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)), a nucleic acid encoding such a polypeptide, or a vector containing such a nucleic acid molecule is administered to a subject in need thereof, to increase bone formation (e.g., increase the rate of bone formation or osteogenesis in the subject).
As used herein, the phrase "affecting myostatin, activin, and/or BMP9 signaling" means changing the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla). In some embodiments, a polypeptide including an extracellular ActRlla variant described herein reduces or inhibits the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla). As described herein, a polypeptide of the invention including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 - 72 (e.g., SEQ ID NOs: 6-72)) may have weak binding affinity to BMP9 (e.g., KD of 200 pM or higher).
As used herein, the term "vascular complication" refers to a vascular disorder or any damage to the blood vessels, such as damage to the blood vessel walls. Damage to the blood vessel walls may cause an increase in vascular permeability or leakage. The term "vascular permeability or leakage" refers to the capacity of the blood vessel walls to allow the flow of small molecules, proteins, and cells in and out of blood vessels. An increase in vascular permeability or leakage may be caused by an increase in the gaps (e.g., an increase in the size and/or number of the gaps) between endothelial cells that line the blood vessel walls and/or thinning of the blood vessel walls.
As used herein, the term "polypeptide" describes a single polymer in which the monomers are amino acid residues which are covalently conjugated together through amide bonds. A polypeptide is intended to encompass any amino acid sequence, either naturally occurring, recombinant, or synthetically produced.
As used herein, the term "homodimer" refers to a molecular construct formed by two identical macromolecules, such as proteins or nucleic acids. The two identical monomers may form a homodimer by covalent bonds or non-covalent bonds. For example, an Fc domain may be a homodimer of two Fc domain monomers if the two Fc domain monomers contain the same sequence. In another example, a polypeptide described herein including an extracellular ActRlla variant fused to an Fc domain monomer may form a homodimer through the interaction of two Fc domain monomers, which form an Fc domain in the homodimer.
As used herein, the term "heterodimer" refers to a molecular construct formed by two different macromolecules, such as proteins or nucleic acids. The two monomers may form a heterodimer by covalent bonds or non-covalent bonds. For example, a polypeptide described herein including an extracellular ActRlla variant fused to an Fc domain monomer may form a heterodimer through the interaction of two Fc domain monomers, each fused to a different ActRlla variant, which form an Fc domain in the heterodimer.
As used herein, the term "host cell" refers to a vehicle that includes the necessary cellular components, e.g., organelles, needed to express proteins from their corresponding nucleic acids. The nucleic acids are typically included in nucleic acid vectors that can be introduced into the host cell by conventional techniques known in the art (transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, etc.). A host cell may be a prokaryotic cell, e.g., a bacterial cell, or a eukaryotic cell, e.g., a mammalian cell (e.g., a CHO cell or a HEK293 cell).
As used herein, the term "therapeutically effective amount" refers an amount of a polypeptide, nucleic acid, or vector of the invention or a pharmaceutical composition containing a polypeptide, nucleic acid, or vector of the invention effective in achieving the desired therapeutic effect in treating a patient having a disease, such as -osteoporosis, or a condition involving bone damage, e.g., primary
osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet- related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss. The term "therapeutically effective amount" also refers an amount of a polypeptide, nucleic acid, or vector of the invention or a pharmaceutical composition containing a polypeptide, nucleic acid, or vector of the invention effective in achieving the desired therapeutic effect in treating a patient having a disease, such as a disease or condition involving bone damage. In particular, the therapeutically effective amount of the polypeptide, nucleic acid, or vector avoids adverse side effects.
As used herein, the term "pharmaceutical composition" refers to a medicinal or pharmaceutical formulation that includes an active ingredient as well as excipients and diluents to enable the active ingredient suitable for the method of administration. The pharmaceutical composition of the present invention includes pharmaceutically acceptable components that are compatible with the polypeptide, nucleic acid, or vector. The pharmaceutical composition may be in tablet or capsule form for oral administration or in aqueous form for intravenous or subcutaneous administration.
As used herein, the term "pharmaceutically acceptable carrier or excipient" refers to an excipient or diluent in a pharmaceutical composition. The pharmaceutically acceptable carrier must be compatible with the other ingredients of the formulation and not deleterious to the recipient. In the present invention, the pharmaceutically acceptable carrier or excipient must provide adequate pharmaceutical stability to the polypeptide including an extracellular ActRlla variant, the nucleic acid molecule(s) encoding the polypeptide, or a vector containing such nucleic acid molecule(s). The nature of the carrier or excipient differs with the mode of administration. For example, for intravenous administration, an aqueous solution carrier is generally used; for oral administration, a solid carrier is preferred.
As used herein, the term "treating and/or preventing" refers to the treatment and/or prevention of a disease, e.g., a bone disease or condition (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), using methods and compositions of the invention. Generally, treating a bone disease or condition occurs after a subject has developed the bone disease or condition and/or is already diagnosed with the bone disease or condition. Preventing a bone disease or condition refers to steps or procedures taken when a subject is at risk of developing the bone disease or condition. The subject may show signs or mild symptoms that are judged by a physician to be indications or risk factors for developing the bone disease or condition or have a family history or genetic predisposition of developing the bone disease or condition, but has not yet developed the disease.
As used herein, the term "subject" refers to a mammal, e.g., preferably a human. Mammals include, but are not limited to, humans and domestic and farm animals, such as monkeys (e.g., a cynomolgus monkey), mice, dogs, cats, horses, and cows, etc.
DESCRIPTION OF THE DRAWINGS
FIG. 1 is a sequence alignment showing the wild-type sequences of extracellular ActRlla and ActRllb and the amino acid substitutions in ActRlla variants. DETAILED DESCRIPTION OF THE INVENTION
The invention features polypeptides that include an extracellular activin receptor type Ma (ActRlla) variant. In some embodiments, a polypeptide of the invention includes an extracellular ActRlla variant fused to a moiety (e.g., Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin). A polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may also form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers. The ActRlla variants described herein have weak binding affinity or no binding affinity to bone morphogenetic protein 9 (BMP9) compared to activins and myostatin. The invention also includes methods of treating diseases and conditions involving bone damage by increasing bone mineral density or bone formation or affecting myostatin, activin, and/or BMP9 signaling in a subject by administering to the subject a polypeptide including an extracellular ActRlla variant described herein. I. Extracellular activin receptor type Ma (ActRlla) variants
Activin type II receptors are single transmembrane domain receptors that modulate signals for ligands in the transforming growth factor β (TGF-β) superfamily. Ligands in the TGF-β superfamily are involved in a host of physiological processes, such as muscle growth, vascular growth, cell differentiation, homeostasis, and osteogenesis. Examples of ligands in the TGF-β superfamily include, e.g., activin, inhibin, growth differentiation factors (GDFs) (e.g., GDF8, also known as myostatin), and bone morphogenetic proteins (BMPs) (e.g., BMP9). Activins are expressed abundantly in bone tissues and regulate bone formation by controlling both osteoblast and osteoclast functions. Activin has been reported to be upregulated in bone disease and inhibit osteoblast activity. Myostatin is also implicated in bone homeostasis through increasing osteogenesis and inhibiting osteoblast activity. These data suggest that activin receptor ligands (e.g., activin and myostatin), promote bone resorption, which could lead to diseases and conditions involving bone damage, such as primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss. Methods that reduce or inhibit this signaling could, therefore, be used in the treatment of diseases and conditions involving bone damage.
There exist two types of activin type II receptors: ActRlla and ActRllb. Studies have shown that BMP9 binds ActRllb with about 300-fold higher binding affinity than ActRlla (see, e.g., Townson et al., J. Biol. Chem. 287:27313, 2012). ActRlla is known to have a longer half-life compared to ActRllb. The present invention describes extracellular ActRlla variants that are constructed by introducing amino acid residues of ActRllb to ActRlla, with the goal of imparting physiological properties conferred by ActRllb, while also maintaining beneficial physiological and pharmacokinetic properties of ActRlla. The optimum peptides confer significant increases in bone mineral density, while retaining longer serum half-life and low binding-affinity to BMP9, for example. The preferred ActRlla variants also exhibit improved binding to activins and/or myostatin compared to wild-type ActRlla, which allows them to compete with endogenous activin receptors for ligand binding and reduce or inhibit endogenous activin receptor signaling. These variants can be used to treat disorders in which activin receptor signaling is elevated, such bone disease, leading to a reduction in bone resorption or osteoclast activity, and in increase in bone formation, bone mineral density, or bone strength. In some embodiments, amino acid substitutions may be introduced to an extracellular ActRlla variant to reduce or remove the binding affinity of the variant to BMP9. The wild- type amino acid sequences of the extracellular portions of human ActRlla and ActRllb are shown below.
Human ActRlla, extracellular portion (SEQ ID NO: 73):
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIVKQGC WLDDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS Human ActRllb, extracellular portion (SEQ ID NO: 74):
GRGEAETRECIYYNANWELERTNQSGLERCEGEQDKRLHCYASWRNSSGTIELVKKGCWL DDFNCYDRQECVATEENPQVYFCCCEGNFCNERFTHLPEAGGPEVTYEPPPTAPT
Polypeptides described herein include an extracellular ActRlla variant having at least one amino acid substitution relative to the wild-type extracellular ActRlla having the sequence of SEQ ID NO: 73 or the extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96. Possible amino acid substitutions at 27 different positions may be introduced to an extracellular ActRlla variant (Table 1 ). In some embodiments, an extracellular ActRlla variant may have at least 85% (e.g., at least 85%, 87%, 90%, 92%, 95%, 97%, or greater) amino acid sequence identity to the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73). An extracellular ActRlla variant may have one or more (e.g., 1 -27, 1 -25, 1 -23, 1 -21 , 1 -19, 1 -17, 1 -15, 1 -13, 1 -1 1 , 1 -9, 1 -7, 1 -5, 1 -3, or 1 -2; e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 1 7, 18, 19, 20, 21 , 22, 23, 24, 25, 26, or 27) amino acid substitutions relative the sequence of a wild-type extracellular ActRlla (SEQ ID NO: 73). In some embodiments, an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of SEQ ID NO: 1 ) may include amino acid substitutions at all of the 27 positions as listed in Table 1 . In some embodiments, an extracellular ActRlla variant may include amino acid substitutions at a number of positions, e.g., at 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, or 26 out of the 27 positions, as listed in Table 1 .
Amino acid substitutions can worsen or improve the activity and/or binding affinity of the ActRlla variants of the invention. To maintain polypeptide function, it is important that the lysine (K) at position Xi7 in the sequences shown in Tables 1 and 2 (SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) be retained. Substitutions at that position can lead to a loss of activity. For example, an ActRlla variant having the sequence
GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIVAKGCWLDDFNCYD
RTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 150) has reduced activity in vivo, indicating that the substitution of alanine (A) for lysine (K) at X17 is not tolerated. ActRlla variants of the invention, including variants in Tables 1 and 2 (e.g., SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72), therefore, retain amino acid K at position X17.
The ActRlla variants of the invention preferably have reduced, weak, or no substantial binding to
BMP9. BMP9 binding is reduced in ActRlla variants containing the amino acid sequence TEEN at positions X23, X24, X25, and X26, as well as in variants that maintain the amino acid K at position X24 and have the amino acid sequence TKEN at positions X23, X24, X25, and X26. The sequences TEEN and TKEN can be employed interchangeably in the ActRlla variants (e.g., the variants in Tables 1 and 2, e.g., SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) of the invention to provide reduced BMP9 binding.
The ActRlla variants of the invention may further include a C-terminal extension (e.g., additional amino acids at the C-terminus). The C-terminal extension can add one or more additional amino acids at the C-terminus (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids) to any of the variants shown in Tables 1 and 2 (e.g., SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)). One potential C-terminal extension that can be included in the ActRlla variants of the invention is amino acid sequence NP. For example, the sequence including the C-terminal extension is SEQ ID NO: 71 (e.g., SEQ ID NO: 69 with a C- terminal extension of NP). Another exemplary C-terminal extension that can be included in the ActRlla variants of the invention is amino acid sequence NPVTPK (SEQ ID NO: 155). For example, the sequence including the C-terminal extension is SEQ ID NO: 72 (e.g., SEQ ID NO: 69 with a C-terminal extension of NPVTPK).
Amino acid substitutions in an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -5
In some embodiments of the extracellular ActRlla variant having the sequence of SEQ ID NO: 2, X3 is E, X6 is R, Xi i is D, X12 is K, X13 is R, Xi6 is K or R, X17 is K, X19 is W, X20 is L, X21 is D, and X22 is I or F. In some embodiments of the extracellular ActRlla variant having the sequence of SEQ ID NO: 1 or 2, Xi7 is K. In some embodiments of the extracellular ActRlla variant having the sequence of SEQ ID NOs: 1 -3, Xi7 is K, X23 is T, X24 is E, X25 is E, and X26 is N. In some embodiments of the extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -5, X17 is K, X23 is T, X24 is K, X25 is E, and X26 is N.
In some embodiments, a polypeptide described herein includes an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 6-72 (Table 2).
Table 2. Extracellular ActRlla variants having the sequences of SEQ ID NOs: 6-72
SEQ ID NO Amino Acid Sequence
18 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
19 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
20 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
21 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
22 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
23 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
24 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
25 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
26 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
27 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
28 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
29 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
30 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
31 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
32 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
33 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
34 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS SEQ ID NO Amino Acid Sequence
35 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
36 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
37 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
38 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
39 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
40 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
41 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
42 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
43 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGC
WLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
44 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
45 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
46 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
47 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
48 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
49 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
50 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS SEQ ID NO Amino Acid Sequence
51 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
52 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
53 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
54 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
55 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
56 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
57 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
58 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
59 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
60 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
61 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
62 GAILGRSETQECLFYNANWELDRTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
63 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETKENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
64 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDINCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
65 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWKNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
66 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCFATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
67 GAILGRSETQECLFYNANWELDRTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS SEQ ID NO Amino Acid Sequence
69 GAILGRSETQECLFYNANWELERTNQTGVEPCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
69 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
70 GAILGRSETQECLYYNANWELERTNQTGVERCEGEQDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS
71 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTSN
P
72 GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIV
KKGCWLDDFNCYDRTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTSN PVTPK
In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) has amino acid K at position X17. Altering the amino acid at position X17 can result in reduced activity. For example, an ActRlla variant having the sequence
GAILGRSETQECLFYNANWELERTNQTGVERCEGEKDKRLHCYATWRNISGSIEIVAKGCWLDDFNCYD RTDCVETEENPQVYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 150) has reduced activity in vivo, indicating that the substitution of A for K at X17 is not tolerated.
In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) with the sequence TEEN at positions X23, X24, X25, and X26 can have a substitution of the amino acid K for the amino acid E at position X24. In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) with the sequence TKEN at positions X23, X24, X25, and X26 can have a substitution of the amino acid E for the amino acid K at position X24. Polypeptides having the sequence TEEN or TKEN at positions X23, X24, X25, and X26 have reduced or weak binding to BMP9.
In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant (e.g., any one of SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)) may further include a C-terminal extension (e.g., additional amino acids at the C-terminus). In some embodiments, the C-terminal extension is amino acid sequence NP. For example, the sequence including the C-terminal extension is SEQ ID NO: 71 (e.g., SEQ ID NO: 69 with a C-terminal extension of NP). In some embodiments, the C-terminal extension is amino acid sequence NPVTPK (SEQ ID NO: 155). For example, the sequence including the C-terminal extension is SEQ ID NO: 72 (e.g., SEQ ID NO: 69 with a C-terminal extension of NPVTPK). The C-terminal extension can add one or more additional amino acids at the C-terminus (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids).
In some embodiments, a polypeptide of the invention including an extracellular ActRlla variant may further include a moiety (e.g., Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin), which may be fused to the N- or C-terminus (e.g., C- terminus) of the extracellular ActRlla variant by way of a linker. In some embodiments, the moiety increases the stability or improves the pharmacokinetic properties of the polypeptide. A polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers, which combine to form an Fc domain in the dimer.
In some embodiments, an extracellular ActRlla variant described herein does not have the sequence of any one of SEQ ID NOs: 76-96 shown in Table 3 below.
Table 3. Excluded Extracellular ActRlla Variants.
SEQ ID NO Amino Acid Sequence
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
88
KQGCWLKDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
89
KQGCWLRDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
90
KQGCWLADINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
91
KQGCWLFDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
92
KQGCWLGDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
93
KQGCWLMDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
94
KQGCWLNDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
95
KQGCWLIDINCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
GAILGRSETQECLFFNANWEKDRTNQTGVEPCYGDKDKRRHCFATWKNISGSIEIV
96
KQGCWLDDANCYDRTDCVEKKDSPEVYFCCCEGNMCNEKFSYFPEMEVTQPTS
Furthermore, in some embodiments, a polypeptide described herein has a serum half-life of at least 7 days in humans. The polypeptide may bind to bone morphogenetic protein 9 (BMP9) with a KD of 200 pM or higher. The polypeptide may bind to activin A with a KD of 10 pM or higher. In some embodiments, the polypeptide does not bind to BMP9 or activin A. In some embodiments, the polypeptide binds to activin and/or myostatin and exhibits reduced (e.g., weak) binding to BMP9. In some embodiments, the polypeptide that has reduced or weak binding to BMP9 has the sequence TEEN or TKEN at positions X23, X24, X25, and X26.
Additionally, in some embodiments, the polypeptide may bind to human BMP9 with a KD of about 200 pM or higher (e.g., a KD of about 200, 300, 400, 500, 600, 700, 800, or 900 pM or higher, e.g., a KD of about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, or 50 nM or higher, e.g., a KD of between about 200 pM and about 50 nM). In some embodiments, the polypeptide does not substantially bind to human BMP9. In some embodiments, the polypeptide may bind to human activin A with a KD of about 800 pM or less (e.g., a KD of about 800, 700, 600, 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 pM or less, e.g., a KD of between about 800 pM and about 200 pM). In some embodiments, the polypeptide may bind to human activin B with a KD of 800 pM or less (e.g., a KD of about 800, 700, 600, 500, 400, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 pM or less, e.g., a KD of between about 800 pM and about 200 pM) The polypeptide may also bind to growth and differentiation factor 1 1 (GDF-1 1 ) with a KD of approximately 5 pM or higher (e.g., a KD of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 1 05, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 1 70, 175, 180, 185, 190, 195, or 200 pM or higher).
II. Fc domains
In some embodiments, a polypeptide described herein may include an extracellular ActRlla variant fused to an Fc domain monomer of an immunoglobulin or a fragment of an Fc domain to increase the serum half-life of the polypeptide. A polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may form a dimer (e.g., homodimer or heterodimer) through the interaction between two Fc domain monomers, which form an Fc domain in the dimer. As conventionally known in the art, an Fc domain is the protein structure that is found at the C-terminus of an immunoglobulin. An Fc domain includes two Fc domain monomers that are dimerized by the interaction between the CH3 antibody constant domains. A wild-type Fc domain forms the minimum structure that binds to an Fc receptor, e.g., FcyRI, FcyRlla, FcyRllb, FcyRllla, FcyRlllb, FcyRIV. In some embodiments, an Fc domain may be mutated to lack effector functions, typical of a "dead" Fc domain. For example, an Fc domain may include specific amino acid substitutions that are known to minimize the interaction between the Fc domain and an Fey receptor. In some embodiments, an Fc domain is from an lgG1 antibody and includes amino acid substitutions L234A, L235A, and G237A. In some embodiments, an Fc domain is from an lgG1 antibody and includes amino acid substitutions D265A, K322A, and N434A. The aforementioned amino acid positions are defined according to Kabat (Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1 991 )). The Kabat numbering of amino acid residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a "standard" Kabat numbered sequence. Furthermore, in some embodiments, an Fc domain does not induce any immune system-related response. For example, the Fc domain in a dimer of a polypeptide including an extracellular ActRlla variant fused to an Fc domain monomer may be modified to reduce the interaction or binding between the Fc domain and an Fey receptor. The sequence of an Fc domain monomer that may be fused to an extracellular ActRlla variant is shown below (SEQ ID NO: 97):
THTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHN AKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPVPIEKTISKAKGQPREPQVYTL PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGPFFLYSKLTVDKS RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
In some embodiments, an Fc domain is from an lgG1 antibody and includes amino acid substitutions L12A, L13A, and G15A, relative to the sequence of SEQ ID NO: 97. In some embodiments, an Fc domain is from an lgG1 antibody and includes amino acid substitutions D43A, K100A, and N212A, relative to the sequence of SEQ ID NO: 97. In some embodiments, an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused to the N- or C-terminus of an Fc domain monomer (e.g., SEQ ID NO: 97) through conventional genetic or chemical means, e.g., chemical conjugation. If desired, a linker (e.g., a spacer) can be inserted between the extracellular ActRlla variant and the Fc domain monomer. The Fc domain monomer can be fused to the N- or C-terminus (e.g., C-terminus) of the extracellular ActRlla variant. In some embodiments, a polypeptide described herein may include an extracellular ActRlla variant fused to an Fc domain. In some embodiments, the Fc domain contains one or more amino acid substitutions that reduce or inhibit Fc domain dimerization. In some embodiments, the Fc domain contains a hinge domain. The Fc domain can be of immunoglobulin antibody isotype IgG, IgE, IgM, IgA, or IgD. Additionally, the Fc domain can be an IgG subtype (e.g., lgG1 , lgG2a, lgG2b, lgG3, or lgG4). The Fc domain can also be a non-naturally occurring Fc domain, e.g., a recombinant Fc domain.
Methods of engineering Fc domains that have reduced dimerization are known in the art. In some embodiments, one or more amino acids with large side-chains (e.g., tyrosine or tryptophan) may be introduced to the CH3-CH3 dimer interface to hinder dimer formation due to steric clash. In other embodiments, one or more amino acids with small side-chains (e.g., alanine, valine, or threonine) may be introduced to the CH3-CH3 dimer interface to remove favorable interactions. Methods of introducing amino acids with large or small side-chains in the CH3 domain are described in, e.g., Ying et al. (J Biol Chem. 287:19399-19408, 2012), U.S. Patent Publication No. 2006/0074225, U.S. Patent Nos. 8,216,805 and 5,731 ,1 68, Ridgway et al. {Protein Eng. 9:617-612, 1996), Atwell et al. (J Mol Biol. 270:26-35, 1997), and Merchant et al. (Nat Biotechnol. 16:677-681 , 1998), all of which are incorporated herein by reference in their entireties.
In yet other embodiments, one or more amino acid residues in the CH3 domain that make up the CH3-CH3 interface between two Fc domains are replaced with positively-charged amino acid residues (e.g., lysine, arginine, or histidine) or negatively-charged amino acid residues (e.g., aspartic acid or glutamic acid) such that the interaction becomes electrostatically unfavorable depending on the specific charged amino acids introduced. Methods of introducing charged amino acids in the CH3 domain to disfavor or prevent dimer formation are described in, e.g., Ying et al. (J Biol Chem. 287:19399-19408, 2012), U.S. Patent Publication Nos. 2006/0074225, 2012/0244578, and 2014/00241 1 1 , all of which are incorporated herein by reference in their entireties.
In some embodiments of the invention, an Fc domain includes one or more of the following amino acid substitutions:T366W, T366Y, T394W, F405W, Y349T, Y349E, Y349V, L351 T, L351 H, L351 N, L352K, P353S, S354D, D356K, D356R, D356S, E357K, E357R, E357Q, S364A, T366E, L368T, L368Y, L368E, K370E, K370D, K370Q, K392E, K392D, T394N, P395N, P396T, V397T, V397Q, L398T, D399K, D399R, D399N, F405T, F405H, F405R, Y407T, Y407H, Y407I, K409E, K409D, K409T, and K409I, relative to the sequence of human lgG1 . In one particular embodiment, an Fc domain includes the amino acid substitution T366W, relative to the sequence of human lgG1 . The sequence of wild-type Fc domain is shown in SEQ ID NO: 151 .
III. Albumin-binding peptide
In some embodiments, a polypeptide described herein may include an extracellular ActRlla variant fused to a serum protein-binding peptide. Binding to serum protein peptides can improve the pharmacokinetics of protein pharmaceuticals.
As one example, albumin-binding peptides that can be used in the methods and compositions described here are generally known in the art. In one embodiment, the albumin binding peptide includes the sequence DICLPRWGCLW (SEQ ID NO: 1 52). In the present invention, albumin-binding peptides may be joined to the N- or C-terminus (e.g., C- terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant. In some embodiments, an albumin-binding peptide is joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
In some embodiments, an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused to the N- or C-terminus of albumin-binding peptide (e.g., SEQ ID NO: 152) through conventional genetic or chemical means, e.g., chemical conjugation. If desired, a linker (e.g., a spacer) can be inserted between the extracellular ActRlla variant and the albumin-binding peptide. Without being bound to a theory, it is expected that inclusion of an albumin-binding peptide in an extracellular ActRlla variant described herein may lead to prolonged retention of the therapeutic protein through its binding to serum albumin. IV. Fibronectin domain
In some embodiments, a polypeptide described herein may include an extracellular ActRlla variant fused to fibronectin domains. Binding to fibronectin domains can improve the pharmacokinetics of protein pharmaceuticals.
Fibronectin domain is a high molecular weight glycoprotein of the extracellular matrix, or a fragment thereof, that binds to, e.g., membrane-spanning receptor proteins such as integrins and extracellular matrix components such as collagens and fibrins. In some embodiments of the present invention, a fibronectin domain is joined to the N- or C-terminus (e.g., C-terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant. A fibronectin domain can be joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
As one example, fibronectin domains that can be used in the methods and compositions described here are generally known in the art. In one embodiment, the fibronectin domain is a fibronectin type III domain (SEQ ID NO: 153) having amino acids 610-702 of the sequence of UniProt ID NO:
P02751 . In another embodiment, the fibronectin domain is an adnectin protein.
In some embodiments, an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused to the N- or C-terminus of a fibronectin domain (e.g., SEQ ID NO: 153) through conventional genetic or chemical means, e.g., chemical conjugation. If desired, a linker (e.g., a spacer) can be inserted between the extracellular ActRlla variant and the fibronectin domain. Without being bound to a theory, it is expected that inclusion of a fibronectin domain in an extracellular ActRlla variant described herein may lead to prolonged retention of the therapeutic protein through its binding to integrins and extracellular matrix components such as collagens and fibrins. V. Serum albumin
In some embodiments, a polypeptide described herein may include an extracellular ActRlla variant fused to serum albumin. Binding to serum albumins can improve the pharmacokinetics of protein pharmaceuticals.
Serum albumin is a globular protein that is the most abundant blood protein in mammals. Serum albumin is produced in the liver and constitutes about half of the blood serum proteins. It is monomeric and soluble in the blood. Some of the most crucial functions of serum albumin include transporting hormones, fatty acids, and other proteins in the body, buffering pH, and maintaining osmotic pressure needed for proper distribution of bodily fluids between blood vessels and body tissues. In preferred embodiments, serum albumin is human serum albumin. In some embodiments of the present invention, a human serum albumin is joined to the N- or C-terminus (e.g., C-terminus) of an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) to increase the serum half-life of the extracellular ActRlla variant. A human serum albumin can be joined, either directly or through a linker, to the N- or C-terminus of an extracellular ActRlla variant.
As one example, serum albumins that can be used in the methods and compositions described herein are generally known in the art. In one embodiment, the serum albumin includes the sequence of UniProt ID NO: P02768 (SEQ ID NO: 154).
In some embodiments, an extracellular ActRlla variant described herein (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be fused to the N- or C-terminus of a human serum albumin (e.g., SEQ ID NO: 1 54) through conventional genetic or chemical means, e.g., chemical conjugation. If desired, a linker (e.g., a spacer) can be inserted between the extracellular ActRlla variant and the serum albumin. Without being bound to a theory, it is expected that inclusion of a serum albumin in an extracellular ActRlla variant described herein may lead to prolonged retention of the therapeutic protein.
VI. Linkers
A polypeptide described herein may include an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having a sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety by way of a linker. In some embodiments, the moiety increases stability of the polypeptide.
Exemplary moieties include an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a human serum albumin. In the present invention, a linker between a moiety (e.g., an Fc domain monomer (e.g., the sequence of SEQ ID NO: 97), a wild-type Fc domain (e.g., SEQ ID NO: 151 ), an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce
dimerization), an albumin-binding peptide (e.g., SEQ ID NO: 152), a fibronectin domain (e.g., SEQ ID NO: 153), or a human serum albumin (e.g., SEQ ID NO: 1 54)) and an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6- 72)), can be an amino acid spacer including 1 -200 amino acids. Suitable peptide spacers are known in the art, and include, for example, peptide linkers containing flexible amino acid residues such as glycine, alanine, and serine. In some embodiments, a spacer can contain motifs, e.g., multiple or repeating motifs, of GA, GS, GG, GGA, GGS, GGG, GGGA (SEQ ID NO: 98), GGGS (SEQ ID NO: 99), GGGG (SEQ ID NO: 100), GGGGA (SEQ ID NO: 101 ), GGGGS (SEQ ID NO: 102), GGGGG (SEQ ID NO: 103), GGAG (SEQ ID NO: 1 04), GGSG (SEQ ID NO: 105), AGGG (SEQ ID NO: 1 06), or SGGG (SEQ ID NO: 107). In some embodiments, a spacer can contain 2 to 12 amino acids including motifs of GA or GS, e.g., GA, GS, GAGA (SEQ ID NO: 1 08), GSGS (SEQ ID NO: 109), GAGAGA (SEQ ID NO: 1 1 0), GSGSGS (SEQ ID NO: 1 1 1 ), GAGAGAGA (SEQ ID NO: 1 12), GSGSGSGS (SEQ ID NO: 1 13),
GAGAGAGAGA (SEQ ID NO: 1 14), GSGSGSGSGS (SEQ ID NO: 1 15), GAGAGAGAGAGA (SEQ ID NO: 1 16), and GSGSGSGSGSGS (SEQ ID NO: 1 17). In some embodiments, a spacer can contain 3 to 12 amino acids including motifs of GGA or GGS, e.g., GGA, GGS, GGAGGA (SEQ ID NO: 1 1 8), GGSGGS (SEQ ID NO: 1 19), GGAGGAGGA (SEQ ID NO: 120), GGSGGSGGS (SEQ ID NO: 121 ), GGAGGAGGAGGA (SEQ ID NO: 122), and GGSGGSGGSGGS (SEQ ID NO: 123). In yet some embodiments, a spacer can contain 4 to 12 amino acids including motifs of GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), e.g., GGAG (SEQ ID NO: 104), GGSG (SEQ ID NO: 1 05), GGAGGGAG (SEQ ID NO: 124), GGSGGGSG (SEQ ID NO: 125), GGAGGGAGGGAG (SEQ ID NO: 126), and
GGSGGGSGGGSG (SEQ ID NO: 127). In some embodiments, a spacer can contain motifs of GGGGA (SEQ ID NO: 101 ) or GGGGS (SEQ ID NO: 102), e.g., GGGGAGGGGAGGGGA (SEQ ID NO: 128) and GGGGSGGGGSGGGGS (SEQ ID NO: 129). In some embodiments of the invention, an amino acid spacer between a moiety (e.g., an Fc domain monomer, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin) and an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be GGG, GGGA (SEQ ID NO: 98), GGGG (SEQ ID NO: 100), GGGAG (SEQ ID NO: 130), GGGAGG (SEQ ID NO: 131 ), or GGGAGGG (SEQ ID NO: 132).
In some embodiments, a spacer can also contain amino acids other than glycine, alanine, and serine, e.g., AAAL (SEQ ID NO: 133), AAAK (SEQ ID NO: 134), AAAR (SEQ ID NO: 135),
EGKSSGSGSESKST (SEQ ID NO: 136), GSAGSAAGSGEF (SEQ ID NO: 137), AEAAAKEAAAKA (SEQ ID NO: 138), KESGSVSSEQLAQFRSLD (SEQ ID NO: 139), G ENLYFQSGG (SEQ ID NO: 140), SACYCELS (SEQ ID NO: 141 ), RSIAT (SEQ ID NO: 142), RPACKIPNDLKQKVMNH (SEQ ID NO: 143), GGSAGGSGSGSSGGSSGASGTGTAGGTGSGSGTGSG (SEQ ID NO: 144), AAANSSIDLISVPVDSR (SEQ ID NO: 145), or GGSGGGSEGGGSEGGGSEGGGSEGGGSEGGGSGGGS (SEQ ID NO: 146). In some embodiments, a spacer can contain motifs, e.g., multiple or repeating motifs, of EAAAK (SEQ ID NO: 147). In some embodiments, a spacer can contain motifs, e.g., multiple or repeating motifs, of proline-rich sequences such as (XP)n, in which X may be any amino acid (e.g., A, K, or E) and n is from 1 - 5, and PAPAP(SEQ ID NO: 148).
The length of the peptide spacer and the amino acids used can be adjusted depending on the two protein involved and the degree of flexibility desired in the final protein fusion polypeptide. The length of the spacer can be adjusted to ensure proper protein folding and avoid aggregate formation.
VII. Vectors, host cells, and protein production
The polypeptides of the invention can be produced from a host cell. A host cell refers to a vehicle that includes the necessary cellular components, e.g., organelles, needed to express the polypeptides and fusion polypeptides described herein from their corresponding nucleic acids. The nucleic acids may be included in nucleic acid vectors that can be introduced into the host cell by conventional techniques known in the art (e.g., transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, infection, or the like). The choice of nucleic acid vectors depends in part on the host cells to be used. Generally, preferred host cells are of either eukaryotic (e.g., mammalian) or prokaryotic (e.g., bacterial) origin.
Nucleic acid vector construction and host cells
A nucleic acid sequence encoding the amino acid sequence of a polypeptide of the invention may be prepared by a variety of methods known in the art. These methods include, but are not limited to, oligonucleotide-mediated (or site-directed) mutagenesis and PCR mutagenesis. A nucleic acid molecule encoding a polypeptide of the invention may be obtained using standard techniques, e.g., gene synthesis. Alternatively, a nucleic acid molecule encoding a wild-type extracellular ActRlla may be mutated to include specific amino acid substitutions using standard techniques in the art, e.g., QuikChange™ mutagenesis. Nucleic acid molecules can be synthesized using a nucleotide synthesizer or PCR techniques.
A nucleic acid sequence encoding a polypeptide of the invention may be inserted into a vector capable of replicating and expressing the nucleic acid molecule in prokaryotic or eukaryotic host cells. Many vectors are available in the art and can be used for the purpose of the invention. Each vector may include various components that may be adjusted and optimized for compatibility with the particular host cell. For example, the vector components may include, but are not limited to, an origin of replication, a selection marker gene, a promoter, a ribosome binding site, a signal sequence, the nucleic acid sequence encoding protein of interest, and a transcription termination sequence.
In some embodiments, mammalian cells may be used as host cells for the invention. Examples of mammalian cell types include, but are not limited to, human embryonic kidney (HEK) (e.g., HEK293, HEK 293F), Chinese hamster ovary (CHO), HeLa, COS, PC3, Vera, MC3T3, NSO, Sp2/0, VERY, BHK, MDCK, W138, BT483, Hs578T, HTB2, BT20, T47D, NSO (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7030, and HsS78Bst cells. In some embodiments, E. coli cells may also be used as host cells for the invention. Examples of E. coli strains include, but are not limited to, E. coli 294 (ATCC® 31 ,446), E. coli K 1776 (ATCC® 31 ,537, E. coli BL21 (DE3) (ATCC® BAA- 1025), and E. coli RV308 (ATCC®31 ,608). Different host cells have characteristic and specific mechanisms for the posttranslational processing and modification of protein products (e.g., glycosylation). Appropriate cell lines or host systems may be chosen to ensure the correct modification and processing of the polypeptide expressed. The above-described expression vectors may be introduced into appropriate host cells using conventional techniques in the art, e.g., transformation, transfection, electroporation, calcium phosphate precipitation, and direct microinjection. Once the vectors are introduced into host cells for protein production, host cells are cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. Methods for expression of therapeutic proteins are known in the art, see, for example, Paulina Balbas, Argelia Lorence (eds.) Recombinant Gene Expression: Reviews and Protocols (Methods in Molecular Biology), Humana Press; 2nd ed. 2004 and Vladimir Voynov and Justin A. Caravella (eds.) Therapeutic Proteins: Methods and Protocols (Methods in Molecular Biology) Humana Press; 2nd ed. 2012.
Protein production, recovery, and purification
Host cells used to produce the polypeptides of the invention may be grown in media known in the art and suitable for culturing of the selected host cells. Examples of suitable media for mammalian host cells include Minimal Essential Medium (MEM), Dulbecco's Modified Eagle's Medium (DMEM), Expi293™ Expression Medium, DMEM with supplemented fetal bovine serum (FBS), and RPMI-1 640. Examples of suitable media for bacterial host cells include Luria broth (LB) plus necessary supplements, such as a selection agent, e.g., ampicillin. Host cells are cultured at suitable temperatures, such as from about 20 °C to about 39 °C, e.g., from 25 °C to about 37 °C, preferably 37 °C, and C02 levels, such as 5 to 10%. The pH of the medium is generally from about 6.8 to 7.4, e.g., 7.0, depending mainly on the host organism. If an inducible promoter is used in the expression vector of the invention, protein expression is induced under conditions suitable for the activation of the promoter.
In some embodiments, depending on the expression vector and the host cells used, the expressed protein may be secreted from the host cells (e.g., mammalian host cells) into the cell culture media. Protein recovery may involve filtering the cell culture media to remove cell debris. The proteins may be further purified. A polypeptide of the invention may be purified by any method known in the art of protein purification, for example, by chromatography (e.g., ion exchange, affinity, and size-exclusion column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. For example, the protein can be isolated and purified by appropriately selecting and combining affinity columns such as Protein A column (e.g., POROS Protein A chromatography) with chromatography columns (e.g., POROS HS-50 cation exchange chromatography), filtration, ultra filtration, salting-out and dialysis procedures.
In other embodiments, host cells may be disrupted, e.g., by osmotic shock, sonication, or lysis, to recover the expressed protein. Once the cells are disrupted, cell debris may be removed by
centrifugation or filtration. In some instances, a polypeptide can be conjugated to marker sequences, such as a peptide to facilitate purification. An example of a marker amino acid sequence is a hexa- histidine peptide (His-tag), which binds to nickel-functionalized agarose affinity column with micromolar affinity. Other peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA" tag, which corresponds to an epitope derived from influenza hemagglutinin protein (Wilson et al., Cell 37:767, 1984).
Alternatively, the polypeptides of the invention can be produced by the cells of a subject (e.g., a human), e.g., in the context of gene therapy, by administrating a vector (such as a viral vector (e.g., a retroviral vector, adenoviral vector, poxviral vector (e.g., vaccinia viral vector, such as Modified Vaccinia Ankara (MVA)), adeno-associated viral vector, and alphaviral vector)) containing a nucleic acid molecule encoding the polypeptide of the invention. The vector, once inside a cell of the subject (e.g., by transformation, transfection, electroporation, calcium phosphate precipitation, direct microinjection, infection, etc.) will promote expression of the polypeptide, which is then secreted from the cell. If treatment of a disease or disorder is the desired outcome, no further action may be required. If collection of the protein is desired, blood may be collected from the subject and the protein purified from the blood by methods known in the art.
VIII. Pharmaceutical compositions and preparations
The invention features pharmaceutical compositions that include the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)). In some embodiments, a pharmaceutical composition of the invention includes a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -70 (e.g., SEQ ID NOs: 6-70)) with a C-terminal extension (e.g., 1 , 2, 3, 4, 5, 6, or more additional amino acids) as the therapeutic protein. In some embodiments, a pharmaceutical composition of the invention includes a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety (e.g., a Fc domain monomer, or a dimer thereof, a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one or more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin) as the therapeutic protein. In some embodiments, a pharmaceutical composition of the invention including a polypeptide of the invention may be used in combination with other agents (e.g., therapeutic biologies and/or small molecules) or compositions in a therapy. In addition to a therapeutically effective amount of the polypeptide, the pharmaceutical composition may include one or more pharmaceutically acceptable carriers or excipients, which can be formulated by methods known to those skilled in the art. In some embodiments, a pharmaceutical composition of the invention includes a nucleic acid molecule (DNA or RNA, e.g., mRNA) encoding a polypeptide of the invention, or a vector containing such a nucleic acid molecule.
Acceptable carriers and excipients in the pharmaceutical compositions are nontoxic to recipients at the dosages and concentrations employed. Acceptable carriers and excipients may include buffers such as phosphate, citrate, HEPES, and TAE, antioxidants such as ascorbic acid and methionine, preservatives such as hexamethonium chloride, octadecyldimethylbenzyl ammonium chloride, resorcinol, and benzalkonium chloride, proteins such as human serum albumin, gelatin, dextran, and
immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acids such as glycine, glutamine, histidine, and lysine, and carbohydrates such as glucose, mannose, sucrose, and sorbitol. Pharmaceutical compositions of the invention can be administered parenterally in the form of an injectable formulation. Pharmaceutical compositions for injection can be formulated using a sterile solution or any pharmaceutically acceptable liquid as a vehicle. Pharmaceutically acceptable vehicles include, but are not limited to, sterile water, physiological saline, and cell culture media (e.g., Dulbecco's Modified Eagle Medium (DMEM), a-Modified Eagles Medium (a-MEM), F-12 medium). Formulation methods are known in the art, see e.g., Banga (ed.) Therapeutic Peptides and Proteins: Formulation, Processing and Delivery Systems (3rd ed.) Taylor & Francis Group, CRC Press (2015).
The pharmaceutical compositions of the invention may be prepared in microcapsules, such as hydroxylmethylcellulose or gelatin-microcapsule and poly-(methylmethacrylate) microcapsule. The pharmaceutical compositions of the invention may also be prepared in other drug delivery systems such as liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules. Such techniques are described in Remington: The Science and Practice of Pharmacy 22th edition (2012). The pharmaceutical compositions to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
The pharmaceutical compositions of the invention may also be prepared as a sustained-release formulation. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptides of the invention. Examples of sustained release matrices include polyesters, hydrogels, polyactides, copolymers of L-glutamic acid and γ ethyl-L- glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as LUPRON DEPOT™, and poly-D-(-)-3-hydroxybutyric acid. Some sustained-release formulations enable release of molecules over a few months, e.g., one to six months, while other formulations release pharmaceutical compositions of the invention for shorter time periods, e.g., days to weeks.
The pharmaceutical composition may be formed in a unit dose form as needed. The amount of active component, e.g., a polypeptide of the invention, included in the pharmaceutical preparations is such that a suitable dose within the designated range is provided (e.g., a dose within the range of 0.01 - 100 mg/kg of body weight).
The pharmaceutical composition for gene therapy can be in an acceptable diluent, or can include a slow release matrix in which the gene delivery vehicle is imbedded. If hydrodynamic injection is used as the delivery method, the pharmaceutical composition containing a nucleic acid molecule encoding a polypeptide described herein or a vector (e.g., a viral vector) containing the nucleic acid molecule is delivered rapidly in a large fluid volume intravenously. Vectors that may be used as in vivo gene delivery vehicle include, but are not limited to, retroviral vectors, adenoviral vectors, poxviral vectors (e.g., vaccinia viral vectors, such as Modified Vaccinia Ankara), adeno-associated viral vectors, and alphaviral vectors.
IX. Routes, dosage, and administration
Pharmaceutical compositions that include the polypeptides of the invention as the therapeutic proteins may be formulated for, e.g., intravenous administration, parenteral administration, subcutaneous administration, intramuscular administration, intra-arterial administration, intrathecal administration, or intraperitoneal administration. The pharmaceutical composition may also be formulated for, or administered via, oral, nasal, spray, aerosol, rectal, or vaginal administration. For injectable formulations, various effective pharmaceutical carriers are known in the art. See, e.g., ASHP Handbook on Injectable Drugs, Toissel, 18th ed. (2014).
In some embodiments, a pharmaceutical composition that includes a nucleic acid molecule encoding a polypeptide of the invention or a vector containing such nucleic acid molecule may be administered by way of gene delivery. Methods of gene delivery are well-known to one of skill in the art. Vectors that may be used for in vivo gene delivery and expression include, but are not limited to, retroviral vectors, adenoviral vectors, poxviral vectors (e.g., vaccinia viral vectors, such as Modified Vaccinia Ankara (MVA)), adeno-associated viral vectors, and alphaviral vectors. In some embodiments, mRNA molecules encoding polypeptides of the invention may be administered directly to a subject.
In some embodiments of the present invention, nucleic acid molecules encoding a polypeptide described herein or vectors containing such nucleic acid molecules may be administered using a hydrodynamic injection platform. In the hydrodynamic injection method, a nucleic acid molecule encoding a polypeptide described herein is put under the control of a strong promoter in an engineered plasmid (e.g., a viral plasmid). The plasmid is often delivered rapidly in a large fluid volume intravenously.
Hydrodynamic injection uses controlled hydrodynamic pressure in veins to enhance cell permeability such that the elevated pressure from the rapid injection of the large fluid volume results in fluid and plasmid extravasation from the vein. The expression of the nucleic acid molecule is driven primarily by the liver. In mice, hydrodynamic injection is often performed by injection of the plasmid into the tail vein. In certain embodiments, mRNA molecules encoding a polypeptide described herein may be administered using hydrodynamic injection.
The dosage of the pharmaceutical compositions of the invention depends on factors including the route of administration, the disease to be treated, and physical characteristics, e.g., age, weight, general health, of the subject. A pharmaceutical composition of the invention may include a dosage of a polypeptide of the invention ranging from 0.01 to 500 mg/kg (e.g., 0.01 , 0.1 , 0.2, 0.3, 0.4, 0.5, 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 1 50, 200, 250, 300, 350, 400, 450, or 500 mg/kg) and, in a more specific embodiment, about 0.1 to about 30 mg/kg and, in a more specific embodiment, about 0.3 to about 30 mg/kg. The dosage may be adapted by the physician in accordance with conventional factors such as the extent of the disease and different parameters of the subject.
The pharmaceutical compositions are administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective to result in an improvement or remediation of the symptoms. The pharmaceutical compositions are administered in a variety of dosage forms, e.g., intravenous dosage forms, subcutaneous dosage forms, and oral dosage forms (e.g., ingestible solutions, drug release capsules). Generally, therapeutic proteins are dosed at 0.1 -100 mg/kg, e.g., 1 -50 mg/kg. Pharmaceutical compositions that include a polypeptide of the invention may be administered to a subject in need thereof, for example, one or more times (e.g., 1 -10 times or more) daily, weekly, biweekly, monthly, bimonthly, quarterly, biannually, annually, or as medically necessary. In some embodiments, pharmaceutical compositions that include a polypeptide of the invention may be administered to a subject in need thereof weekly, biweekly, monthly, bimonthly, or quarterly. Dosages may be provided in either a single or multiple dosage regimens. The timing between administrations may decrease as the medical condition improves or increase as the health of the patient declines. X. Methods of treatment
The invention is based on the discovery that substituting amino acids from the extracellular portion of ActRllb into the extracellular portion ActRlla yields ActRlla variants with improved properties. The ActRlla variants generated by introducing residues from ActRllb into ActRlla retain the beneficial properties of ActRlla, such as longer serum half-life and low binding affinity to BMP9, and gain some of the beneficial properties of ActRllb, such as increased binding to activins A and B (see Table 4). These ActRlla variant properties produce a polypeptide that can be used therapeutically to compete with endogenous activin receptors for ligand binding. As the ActRlla variants contain the extracellular portion of the receptor, they are soluble and able to bind to and sequester ligands (e.g., activins A and B, myostatin, GDF1 1 ) without activating intracellular signaling pathways. Therefore, the extracellular ActRlla variants can be used to treat diseases or conditions in which elevated activin signaling has been implicated (e.g., associated with increased expression of activin receptors or activin receptor ligands). For example, activin has been found to be upregulated in bone disease and is known to inhibit osteoblast activity, suggesting that increased activin levels contribute to bone disease. It follows that treatment with a therapeutic agent that binds to activin and reduces its interaction with endogenous receptors could be used to increase bone mineral density and treat subjects with diseases or conditions involving bone damage.
The invention provides compositions and methods of treatment that may be used to increase bone mineral density, increase bone formation, or reduce bone resorption in a subject in need thereof. In some embodiments, the subject may have a disease that results in bone damage (e.g., osteoporosis or osteopenia). In some embodiments, the methods described herein are directed to affecting myostatin, activin, and/or BMP9 signaling in a subject having a disease or condition involving bone damage. In some embodiments, a polypeptide including an extracellular ActRlla variant described herein reduces or inhibits the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla). In some embodiments, affecting myostatin, activin, and/or BMP9 signaling (e.g., reducing or inhibiting the binding of myostatin, activin, and/or BMP9 to their receptors, e.g., ActRlla, ActRllb, and BMPRII (e.g., ActRlla)) results in an increase in the subject's bone mineral density or bone formation, or a decrease in the subject's bone resorption.
In some embodiments, the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be administered to a subject to increase bone mineral density, to increase bone formation, to decrease bone resorption, or to affect myostatin, activin, and/or BMP9 signaling in the subject. In some embodiments, the methods described herein increase bone mineral density of the subject. In some embodiments, the methods described herein do not cause any vascular complications in the subject, such as increased vascular permeability or leakage. In some embodiments of the methods described herein, the subject has a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss).
The invention also includes methods of treating a subject having primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss by administering to the subject a polypeptide described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 - 72 (e.g., SEQ ID NOs: 6-72)). In some embodiments, the primary osteoporosis is age-related or hormone-related osteoporosis (e.g., related to a decline in estrogen). In some embodiments, the secondary osteoporosis is immobilization-induced or glucocorticoid-induced osteoporosis. In some embodiments, the bone cancer is multiple myeloma or the cancer metastasis-related bone loss is caused by multiple myeloma. In some embodiments, the treatment-related bone loss occurs due to treatment with FGF-21 or GLP-1 , treatment with an FGF-21 or GLP-1 containing therapeutic, or treatment of Type-2 diabetes and/or obesity, or due to cancer therapy (e.g., chemotherapy or radiation). In some embodiments, the diet-related bone loss is rickets (e.g., vitamin D deficiency). In some embodiments, the low-gravity related bone loss is lack of load-related bone loss.
In some embodiments, the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be used to prevent the development of a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, osteopetrosis, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss) and/or to treat patients already diagnosed with a disease or condition involving bone damage. Patients who are likely to develop a disease or condition involving bone damage, e.g., individuals with genetic predisposition, family history of bone damage, or low bone mass, may be administered the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) prophylactically, such that the extracellular ActRlla polypeptides may prevent or delay the development of bone damage.
In some embodiments, the polypeptides described herein (e.g., a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) may be administered to a subject to prevent the development of and/or treat patients with a disease or condition involving bone damage (e.g., primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss), or to affect myostatin, activin, and/or BMP9 signaling in the subject (e.g., to reduce or inhibit the binding of activin, myostatin, and/or BMP9 to their receptors). In some embodiments, the methods described herein increase bone mineral density (e.g., increase bone mass). In some embodiments, the methods described herein reduce bone resorption (e.g., reduce bone catabolic activity). In some embodiments, the methods described herein increase bone formation (e.g., increase bone anabolic activity or increase osteogenesis). In some embodiments, the methods described herein increase osteoblast activity or osteoblastogenesis. In some embodiments, the methods described herein decrease osteoclast activity or osteoclastogenesis. In some embodiments, the methods described herein reduce or inhibit the binding of activin and/or myostatin to their receptors. In some embodiments, the methods increase bone formation, increase bone mineral density, or decrease bone resorption of cortical or trabecular bone.
In any of the methods described herein, a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -71 (e.g., SEQ ID NOs: 6-71 )) that further includes a C-terminal extension of one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, or more amino acids) may be used as the therapeutic protein. In any of the methods described herein, a dimer (e.g., homodimer or heterodimer) of a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to an Fc domain monomer may be used as the therapeutic protein. In any of the methods described herein, a polypeptide including an extracellular ActRlla variant (e.g., an extracellular ActRlla variant having the sequence of any one of SEQ ID NOs: 1 -72 (e.g., SEQ ID NOs: 6-72)) fused to a moiety (e.g., a wild-type Fc domain, an Fc domain with amino acid substitutions (e.g., one more substitutions that reduce dimerization), an albumin-binding peptide, a fibronectin domain, or a serum albumin) may be used as the therapeutic protein. Nucleic acids encoding the polypeptides described herein, or vectors containing said nucleic acids can also be administered according to any of the methods described herein. In any of the methods described herein, the polypeptide, nucleic acid, or vector can be administered as part of a pharmaceutical composition.
EXAMPLES
Example 1 - Evaluation of ActRlla variants binding affinity by surface plasmon resonance (SPR)
The Biacore 3000 was used to measure the kinetics of the interactions between the ActRlla variants and the ligands Activin A, Activin B, growth differentiation factor 1 1 (GDF1 1 ), and BMP-9.
ActRlla variants were transiently expressed in HEK293 cells and purified from the conditioned media using Protein-A Sepharose chromatography. The ActRlla variants were immobilized on the chip (CM4 or CM5) with capture antibodies (anti-mouse from GEGE) in flow cells 2-4 to ensure proper orientation. Flow cell 1 was used as a reference cell to subtract any nonspecific binding and bulk effects. HBS-EP+ buffer from GE Healthcare™ was used as a running buffer. Each ligand was run in a concentration series at 40 μΙ/min to avoid mass transport effects. The data was analyzed using Scrubber2 by BioLogic™ Software to calculate the KD of each interaction (Table 4).
Table 4: Comparison of ActRlla variant binding affinity (KD) to various ligands
Activin A (KD) Activin B (KD) GDF-11 (KD) BMP-9 (KD)
(SEQ ID NO: 38)
ActRlla/bA9m3 variant 71 pM 72.5 pM 1 17 pM 1 .2 nM
(SEQ ID NO: 41)
ActRlla/bA9m4 variant 375 pM 254 pM 394 pM 14-20 nM
(SEQ ID NO: 44)
ActRlla/bmaxl variant 232 pM 97 pM 236 pM 5.6 nM
(SEQ ID NO: 70)
ActRlla/bmax2 variant 135 pM 39 pM 1 13 pM 5 nM
(SEQ ID NO: 71)
ActRlla/bmax3 variant 89 pM 43 pM 214 pM 3.3 nM
(SEQ ID NO: 72)
Example 2 - Effect of extracellular Act R I la variants on bone mineral density
Adult male C57/BL6 mice receive either a sham- (SHAM) or castration-surgery (ORX). Both surgery groups are allowed to recover for 14 days post-surgery. All animals are housed in conventional cages with free access to food (regular chow) and water. SHAM and ORX animals are then assigned to either a vehicle-treated group (VEH) or ActRII variant-treated group and receive bi-weekly systemic intraperitoneal administration of vehicle or ActRII variant (10 mg/kg) for 71 d. Body weights are measured twice per week at the time of treatment. Body composition is analyzed at study day 0 then at days 14, 28, 47, and 71 after treatment initiation using the MiniSpec LF50 NMR Analyzer. At study termination date, tissues of interest (muscles, fat depots, and tibias) are surgically removed, weighed, and properly stored for further analysis. At this time, the ORX animals are also examined to confirm complete removal of testes. Cortical morphometry and trabecular structure of the various bones are also evaluated after the experiment termination using micro-computed tomography.
Other Embodiments
While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth.
All publications, patents, and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
Other embodiments are within the following claims.

Claims

CLAIMS What is claimed is:
1 . A polypeptide comprising an extracellular activin receptor type Ma (ActRlla) variant, the variant having a sequence of
GAILG RSETQECLX1X2NANWX3X4X5X6TNQTGVEX7CX8GX9X10X11X12X13X14HCX15ATWX16NISGSI EIV X17X18GCX19X20X21 DX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 1 ), wherein Xi is F or Y; X2 is F or Y; X3 is E or A; X4 is K or L; X5 is D or E; Χβ is R or A; X7 is P or R; Xs is Y or E; X9 is D or E; X10 is K or Q; Xn is D or A; X12 is K or A; X13 is R or A; X14 is R or L; X15 is F or Y; X16 is K, R, or A; X17 is K, A, Y, F, or I; Xi8 is Q or K; X19 is W or A; X20 is L or A; X21 is D, K, R, A, F, G, M, N, or I ; X22 is I, F, or A; X23 is K or T; X24 is K or E; X25 is D or E; X26 is S or N; and X27 is E or Q, and wherein the variant has at least one amino acid substitution relative to a wild-type extracellular ActRlla having the sequence of SEQ ID NO: 73 or an extracellular ActRlla having any one of the sequences of SEQ ID NOs: 76-96.
2. The polypeptide of claim 1 , wherein the variant has a sequence of
GAILG RSETQECLFX2NANWX3X4X5X6TNQTGVEX7CX8GX9KX11 X12X13X14HCX15ATWX16NISGSIEIVX17 X18GCX19X20X21 DX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 2),
3. The polypeptide of claim 1 or 2, wherein the variant has a sequence of
GAILGRSETQECLFX2NANWEX4X5RTNQTGVEX7CX8GX9KDKRX14HCX15ATWX16NISGSIEIVKX18GCWL DDX22NCYDRTDCVEX23X24X25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 3).
4. The polypeptide of any one of claims 1 -3, wherein the variant has a sequence of
GAILGRSETQECLFX2NANWEX4DRTNQTGVEX7CX8GX9KDKRX14HCX15ATWX16NISGSIEIVKX18GCWL DDX22NCYDRTDCVEX23KX25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 4).
5. The polypeptide of any one of claims 1 -4, wherein the variant has a sequence of
GAILGRSETQECLFX2NANWEX4DRTNQTGVEPCX8GX9KDKRX14HCFATWKNISGSIEIVKX18GCWLDDI NCYDRTDCVEX23KX25X26PX27VYFCCCEGNMCNEKFSYFPEMEVTQPTS (SEQ ID NO: 5).
6. The polypeptide of claim 1 , wherein Xi is F.
7. The polypeptide of claim 1 , wherein Xi is Y.
8. The polypeptide of claim 1 , 6, or 7 wherein X10 is K.
9. The polypeptide of claim 1 , 6, or 7 wherein X10 is Q.
10. The polypeptide of any one of claims 1 -9, wherein X2 is F.
1 1 . The polypeptide of any one of claims 1 -9, wherein X2 is or Y.
12. The polypeptide of any one of claims 1 , 2, and 6-1 1 , wherein X3 is E.
13. The polypeptide of any one of claims 1 , 2, and 6-1 1 , wherein X3 is A.
14. The polypeptide of any one of claims 1 -13, wherein X4 is K.
15. The polypeptide of any one of claims 1 -13, wherein X4 is L.
16. The polypeptide of any one of claims 1 , 2, 3, and 6-15, wherein X5 is D.
17. The polypeptide of any one of claims 1 , 2, 3, and 6-15, wherein X5 is E.
18. The polypeptide of any one of claims 1 , 2 and 6-17, wherein Χβ is R.
19. The polypeptide of any one of claims 1 , 2 and 6-17, wherein Χβ is A.
20. The polypeptide of any one of claims 1 -4 and 6-19, wherein X7 is P.
21 . The polypeptide of any one of claims 1 -4 and 6-19, wherein X7 is R.
22. The polypeptide of any one of claims 1 -21 , wherein Xs is Y.
23. The polypeptide of any one of claims 1 -21 , wherein Xs is E.
24. The polypeptide of any one of claims 1 -23, wherein X9 is D.
25. The polypeptide of any one of claims 1 -23, wherein X9 is E.
26. The polypeptide of any one of claims 1 , 2 and 6-25, wherein Xn is D.
27. The polypeptide of any one of claims 1 , 2 and 6-25, wherein Xn is A.
28. The polypeptide of any one of claims 1 , 2 and 6-27, wherein X12 is K.
29. The polypeptide of any one of claims 1 , 2 and 6-27, wherein X12 is A.
30. The polypeptide o' any one o' claims 1 , 2 and 6-29, wherein X13 is R.
31 . The polypeptide o' any one o' claims 1 , 2 and 6-29, wherein X13 is A.
32. The polypeptide o' any one o' claims 1 -31 , wherein X14 is R.
33. The polypeptide o' any one o' claims 1 -31 , wherein X14 is L.
34. The polypeptide o' any one o' claims 1 -4 and 6-33, wherein X15 is F.
35. The polypeptide o' any one o' claims 1 -4 and 6-33, wherein X15 is Y.
36. The polypeptide o' any one o' claims 1 -4 and 6-35, wherein X16 is K.
37. The polypeptide o' any one o' claims 1 -4 and 6-35, wherein X16 is R.
38. The polypeptide o' any one o' claims 1 -4 and 6-35, wherein X16 is A.
39. The polypeptide o' any one o' claims 1 , 2 and 6-38, wherein X17 is K.
40. The polypeptide o' any one o' claims 1 , 2 and 6-38, wherein X17 is A.
41 . The polypeptide o' any one o' claims 1 , 2 and 6-38, wherein X17 is Y.
42. The polypeptide o' any one o' claims 1 , 2 and 6-38, wherein X17 is F.
43. The polypeptide o' any one o' claims 1 , 2 and 6-38, wherein X17 is I.
44. The polypeptide o' any one o' claims 1 -43, wherein Xie is Q.
45. The polypeptide o' any one o' claims 1 -43, wherein Xie is K.
46. The polypeptide o' any one o' claims 1 , 2 and 6-45, wherein X19 is W
47. The polypeptide o' any one o' claims 1 , 2 and 6-45, wherein X19 is A.
48. The polypeptide o' any one o' claims 1 , 2 and 6-47, wherein X20 is L.
49. The polypeptide o' any one o' claims 1 , 2 and 6-47, wherein X20 is A.
50. The polypeptide o' any one o' claims 1 , 2 and 6-49, wherein X21 is D.
51 . The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is K.
52. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is R.
53. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is A.
54. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is F.
55. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is G.
56. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is M.
57. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is N.
58. The polypeptide of any one o' aims 1 , 2 and 6-49, wherein X21 is I.
59. The polypeptide of any one o' aims 1 -4 and 6-58, wherein X22 is I.
60. The polypeptide of any one o' aims 1 -4 and 6-58, wherein X22 is F.
61 . The polypeptide of any one o' aims 1 -4 and 6-58, wherein X22 is A.
62. The polypeptide of any one o' aims 1 -61 , wherein X23 is K.
63. The polypeptide of any one o' aims 1 -61 , wherein X23 is T.
64. The polypeptide of any one o' aims 1 , 2, 3, and 6-63, wherein X24 is K.
65. The polypeptide of any one o' aims 1 , 2, 3, and 6-63, wherein X24 is E.
66. The polypeptide of any one o' aims 1 -65, wherein X25 is D.
67. The polypeptide of any one o' aims 1 -65, wherein X25 is E.
68. The polypeptide of any one o' aims 1 -67, wherein X26 is S.
69. The polypeptide of any one o' aims 1 -67, wherein X26 is N.
70. The polypeptide of any one o' aims 1 -69, wherein X27 is E.
71 . The polypeptide of any one of claims 1 -69, wherein X27 is Q.
72. The polypeptide of any one of claims 1 -71 , wherein X23 is T, X24 is E, X25 is E, and X26 is N.
73. The polypeptide of any one of claims 1 -71 , wherein X23 is T, X24 is K, X25 is E, and X26 is N.
74. The polypeptide of any one of claims 1 -73, wherein X17 is K.
75. The polypeptide of claim 1 , wherein the variant has the sequence of any one of SEQ ID NOs: 6-72
76. The polypeptide of any one of claims 1 -75, wherein the amino acid at position X24 is replaced with the amino acid K.
77. The polypeptide of any one of claims 1 -75, wherein the amino acid at position X24 is replaced with the amino acid E.
78. The polypeptide of any one of claims 1 -77, further comprising a C-terminal extension of one or more amino acids.
79. The polypeptide of claim 78, wherein the C-terminal extension is NP.
80. The polypeptide of claim 78, wherein the C-terminal extension is NPVTPK.
81 . The polypeptide of any one of claims 1 -80, further comprising an Fc domain monomer fused to the C- terminus of the polypeptide by way of a linker.
82. The polypeptide of claim 81 , wherein the Fc domain monomer comprises the sequence of SEQ ID NO: 97.
83. The polypeptide of any one of claims 1 -80, further comprising a wild-type Fc domain fused to the C- terminus of the polypeptide by way of a linker.
84. The polypeptide of claim 83, wherein the wild-type Fc domain comprises the sequence of SEQ ID NO: 151 .
85. The polypeptide of any one of claims 1 -80, further comprising an Fc domain with amino acid substitutions fused to the C-terminus of the polypeptide by way of a linker.
86. The polypeptide of claim 85, wherein the Fc domain does not form a dimer.
87. The polypeptide of any one of claims 1 -80, further comprising an albumin-binding peptide fused to the C-terminus of the polypeptide by way of a linker.
88. The polypeptide of claim 87, wherein the albumin-binding peptide comprises the sequence of SEQ ID NO: 152.
89. The polypeptide of any one of claims 1 -80, further comprising a fibronectin domain fused to the C- terminus of the polypeptide by way of a linker.
90. The polypeptide of claim 89, wherein the fibronectin domain comprises the sequence of SEQ ID NO: 153.
91 . The polypeptide of any one of claims 1 -80, further comprising a human serum albumin fused to the C-terminus of the polypeptide by way of a linker.
92. The polypeptide of claim 91 , wherein the human serum albumin comprises the sequence of SEQ ID NO: 154.
93. The polypeptide of claim 81 or 82, wherein the polypeptide forms a dimer.
94. The polypeptide of any one of claims 81 -93, wherein the linker is an amino acid spacer.
95. The polypeptide of claim 94, wherein the amino acid spacer is GGG, GGGA (SEQ ID NO: 98), GGGG (SEQ ID NO: 100), GGGAG (SEQ ID NO: 130), GGGAGG (SEQ ID NO: 131 ), or GGGAGGG (SEQ ID NO: 132).
96. The polypeptide of any one of claims 1 -95, wherein the polypeptide has a serum half-life of at least 7 days.
97. The polypeptide of any one of claims 1 -96, wherein the polypeptide binds to human bone morphogenetic protein 9 (BMP9) with a KD of 200 pM or higher.
98. The polypeptide of claim 97, wherein the polypeptide binds to activin and/or myostatin and has reduced or weak binding to human BMP9.
99. The polypeptide of claim 97 or 98, wherein the polypeptide does not substantially bind to human BMP9.
100. The polypeptide of any one of claims 1 -99, wherein the polypeptide binds to human activin A with a KD of 800 pM or less.
1 01 . The polypeptide of any one of claims 1 -1 00, wherein the polypeptide binds to human activin B with a KD of 800 pM or less.
1 02. The polypeptide of any one of claims 1 -1 01 , wherein the polypeptide binds to human GDF-1 1 with a KD of 5 pM or higher.
1 03. A nucleic acid molecule encoding a polypeptide of any of claims 1 -1 02.
1 04. A vector comprising the nucleic acid molecule of claim 1 03.
1 05. A host cell that expresses a polypeptide of any one of claims 1 -1 02, wherein the host cell comprises a nucleic acid molecule of claim 1 03 or a vector of claim 1 04, wherein the nucleic acid molecule or vector is expressed in the host cell.
1 06. A method of preparing a polypeptide of any one of claims 1 -1 02, wherein the method comprising: a) providing a host cell comprising a nucleic acid molecule of claim 1 03 or a vector of claim 1 04, and
b) expressing the nucleic acid molecule or vector in the host cell under conditions that allow for the formation of the polypeptide.
1 07. A pharmaceutical composition comprising a polypeptide of any one of claims 1 -1 02, a nucleic acid molecule of claim 1 03, or a vector of claim 1 04, and one or more pharmaceutically acceptable carriers or excipients.
1 08. The pharmaceutical composition of claim 1 07, wherein the polypeptide is in a therapeutically effective amount.
1 09. A method of increasing bone mineral density in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -1 02, a nucleic acid molecule of claim 1 03, a vector of claim 1 04, or a pharmaceutical composition of claim 1 07 or 1 08.
1 1 0. A method of reducing bone resorption in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -1 02, a nucleic acid molecule of claim 1 03, a vector of claim 1 04, or a pharmaceutical composition of claim 1 07 or 1 08.
1 1 1 . A method of increasing bone formation in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -1 02, a nucleic acid molecule of claim 1 03, a vector of claim 1 04, or a pharmaceutical composition of claim 1 07 or 1 08.
1 12. The method of any one of claims 109-1 1 1 , wherein the subject has primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.
1 13. A method of affecting myostatin, activin, and/or BMP9 signaling in a subject having a disease or condition involving bone damage, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
1 14. The method of claims 1 13, wherein the disease or condition is primary osteoporosis, secondary osteoporosis, osteopenia, osteopetrosis, facture, bone cancer or cancer metastasis-related bone loss, Paget's disease, renal osteodystrophy, treatment-related bone loss, diet-related bone loss, bone loss associated with the treatment of obesity, low gravity-related bone loss, or immobility-related bone loss.
1 15. A method of treating a subject having primary osteoporosis, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
1 16. A method of treating a subject having secondary osteoporosis, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 1 08.
1 17. A method of treating a subject having osteopenia, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
1 18. A method of treating a subject having fracture, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
1 19. A method of treating a subject having bone cancer or cancer metastasis-related bone loss, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
120. A method of treating a subject having Paget's disease, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
121 . A method of treating a subject having renal osteodystrophy, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
122. A method of treating a subject having treatment-related bone loss, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 1 08.
123. A method of treating a subject having diet-related bone loss, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 108.
124. A method of treating a subject having low gravity-related bone loss, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 1 08.
125. A method of treating a subject having immobility-related bone loss, comprising administering to the subject a therapeutically effective amount of a polypeptide of any one of claims 1 -102, a nucleic acid molecule of claim 103, a vector of claim 104, or a pharmaceutical composition of claim 107 or 1 08.
126. The method of any one of claims 1 12, 1 14, and 1 15, wherein the primary osteoporosis is age- related osteoporosis.
127. The method of any one of claims 1 12, 1 14, and 1 15, wherein the primary osteoporosis is hormone- related osteoporosis.
128. The method of any one of claims 1 12, 1 14, and 1 16, wherein the secondary osteoporosis is immobilization-induced osteoporosis.
129. The method of any one of claims 1 12, 1 14, and 1 16, wherein the secondary osteoporosis is glucocorticoid-induced osteoporosis.
130. The method of any one of claims 1 12, 1 14, and 1 19, wherein the cancer is multiple myeloma.
131 . The method of any one of claims 1 12, 1 14, and 122, wherein the treatment is FGF-21 treatment.
132. The method of any one of claims 1 12, 1 14, and 122, wherein the treatment is GLP-1 treatment.
133. The method of any one of claims 1 12, 1 14, and 122, wherein the treatment is cancer therapy.
134. The method of any one of claims 1 12, 1 14, and 122, wherein the treatment is treatment for obesity or Type-2 diabetes.
135. The method of any one of claims 1 12, 1 14, and 123, wherein the diet-related bone loss is rickets.
136. The method of any one of claims 109-135, wherein the method increases bone formation in the subject.
137. The method of any one of claims 109-136, wherein the method decreases bone resorption in the subject.
138. The method of any one of claims 109-137, wherein the method increases osteoblast activity or osteoblastogenesis.
139. The method of any one of claims 109-138, wherein the method decreases osteoclast activity or decreases osteoclastogenesis.
140. The method of any one of claims 109-139, wherein the method reduces or inhibits the binding of activin and/or myostatin to their receptors.
141 . The method of any one of claims 109-140, wherein the method does not cause a vascular complication in the subject.
142. The method of claim 141 , wherein the method does not increase vascular permeability or leakage.
143. The method of any one of claims 109-142, wherein the method increases bone mineral density in the subject.
144. The method of any one of claims 109-143, wherein the bone is cortical bone.
145. The method of any one of claims 109-144, wherein the bone is trabecular bone.
146. The method of any one of claims 109-145, wherein the polypeptide, nucleic acid, vector, or pharmaceutical composition is administered in an amount sufficient to increase bone density, reduce bone resorption, reduce the rate of bone resorption, increase bone formation, increase the rate of bone formation, reduce osteoclast activity, increase osteoblast activity, or affect myostatin, activin, and/or BMP9 signaling in the subject.
EP17868622.6A 2016-11-10 2017-11-09 Activin receptor type iia variants and methods of use thereof Pending EP3538123A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662420476P 2016-11-10 2016-11-10
US201762531943P 2017-07-13 2017-07-13
PCT/US2017/060970 WO2018089715A1 (en) 2016-11-10 2017-11-09 Activin receptor type iia variants and methods of use thereof

Publications (2)

Publication Number Publication Date
EP3538123A1 true EP3538123A1 (en) 2019-09-18
EP3538123A4 EP3538123A4 (en) 2020-10-14

Family

ID=62109935

Family Applications (2)

Application Number Title Priority Date Filing Date
EP17870560.4A Pending EP3538128A4 (en) 2016-11-10 2017-11-09 Activin receptor type iia variants and methods of use thereof
EP17868622.6A Pending EP3538123A4 (en) 2016-11-10 2017-11-09 Activin receptor type iia variants and methods of use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP17870560.4A Pending EP3538128A4 (en) 2016-11-10 2017-11-09 Activin receptor type iia variants and methods of use thereof

Country Status (7)

Country Link
US (4) US11013785B2 (en)
EP (2) EP3538128A4 (en)
JP (4) JP7051846B2 (en)
CN (2) CN110430890A (en)
AU (2) AU2017357944A1 (en)
CA (2) CA3043180A1 (en)
WO (2) WO2018089706A2 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110036025B (en) 2016-10-05 2024-03-22 阿塞勒隆制药公司 Variant ActRIIB proteins and uses thereof
CA3043180A1 (en) 2016-11-10 2018-05-17 Keros Therapeutics, Inc. Activin receptor type iia variants and methods of use thereof
AU2018364668A1 (en) * 2017-11-09 2020-06-11 Keros Therapeutics, Inc. Activin receptor type lla variants and methods of use thereof
US11919938B2 (en) * 2017-11-30 2024-03-05 Grifols Diagnostic Solutions Inc. Immunoassays and engineered proteins for monitoring antibody treatments to the immune checkpoint inhibitors PD1 and PD-L1
EP3737406A4 (en) 2018-01-12 2021-11-03 Keros Therapeutics, Inc. Activin receptor type iib variants and methods of use thereof
KR20210006952A (en) * 2018-05-09 2021-01-19 케로스 테라퓨틱스, 인크. Activin receptor type IIA variant and methods of use thereof
IL296394A (en) * 2020-03-20 2022-11-01 Keros Therapeutics Inc Activin receptor type ii chimeras and methods of use thereof
EP4230196A1 (en) 2022-02-21 2023-08-23 Som Innovation Biotech, S.A. Compounds for use in the treatment of dystrophinopathies

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040223966A1 (en) 2002-10-25 2004-11-11 Wolfman Neil M. ActRIIB fusion polypeptides and uses therefor
EP2314617B1 (en) 2004-07-23 2015-06-24 Acceleron Pharma Inc. ActRII receptor polypeptides
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
US8067562B2 (en) 2005-11-01 2011-11-29 Amgen Inc. Isolated nucleic acid molecule comprising the amino acid sequence of SEQ ID NO:1
PT1973559E (en) 2005-11-23 2013-02-19 Acceleron Pharma Inc Activin-actriia antagonists and uses for promoting bone growth
US8101564B2 (en) 2006-05-03 2012-01-24 Trustees Of The University Of Pennsylvania Methods for regulating osteoclast differentiation and bone resorption using LRRc17
ME02335B (en) 2006-12-18 2013-04-30 Acceleron Pharma Inc Activin-actrii antagonists and uses for treating anemia
US8895016B2 (en) 2006-12-18 2014-11-25 Acceleron Pharma, Inc. Antagonists of activin-actriia and uses for increasing red blood cell levels
TWI584815B (en) 2007-02-01 2017-06-01 艾瑟勒朗法瑪公司 Activin-actriia antagonists and uses for treating or preventing breast cancer
TW201627320A (en) 2007-02-02 2016-08-01 艾瑟勒朗法瑪公司 Variants derived from ActRIIB and uses therefor
CA3039330C (en) * 2007-02-09 2021-11-09 Acceleron Pharma Inc. Activin-actriia antagonists and uses for promoting bone growth in cancer patients
US8501678B2 (en) 2007-03-06 2013-08-06 Atara Biotherapeutics, Inc. Variant activin receptor polypeptides and uses thereof
TW201718635A (en) 2007-03-06 2017-06-01 安美基公司 Variant activin receptor polypeptides and uses thereof
WO2013106175A1 (en) 2011-12-19 2013-07-18 Amgen Inc. Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof
WO2009015345A1 (en) 2007-07-25 2009-01-29 Amgen Inc. Pharmaceutical compositions comprising fc fusion proteins
CN103877564A (en) 2007-09-18 2014-06-25 阿塞勒隆制药公司 Activin-actriia antagonists and uses for decreasing or inhibiting fsh secretion
WO2009111793A2 (en) 2008-03-07 2009-09-11 Myoscience, Inc. Subdermal tissue remodeling using myostatin, methods and related systems
KR101871510B1 (en) 2008-06-26 2018-06-26 악셀레론 파마 인코포레이티드 Methods for dosing an activin-actriia antagonist and monitoring of treated patients
NZ590326A (en) * 2008-06-26 2013-05-31 Acceleron Pharma Inc Antagonists of activin-actriia and uses for increasing red blood cell levels
TWI617316B (en) 2008-08-14 2018-03-11 艾瑟勒朗法瑪公司 Use of gdf traps to increase red blood cell levels
US8216997B2 (en) 2008-08-14 2012-07-10 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating anemia using a combination of GDF traps and erythropoietin receptor activators
AR074397A1 (en) 2008-11-26 2011-01-12 Amgen Inc STABILIZED ACTIVINE IIB RECEPTING POLYEPTIDES AND USES OF THE SAME
CA2749544A1 (en) 2009-01-13 2010-07-22 Acceleron Pharma Inc. Methods for increasing adiponectin
CN104840944A (en) 2009-06-08 2015-08-19 阿塞勒隆制药公司 Methods for increasing thermogenic adipocytes
BR122021017187B1 (en) 2009-06-12 2022-07-12 Acceleron Pharma Inc NUCLEIC ACID AND RECOMBINANT POLYPEPTIDE, PHARMACEUTICAL PREPARATION COMPRISING SUCH POLYPEPTIDE AND THERAPEUTIC USES THEREOF
RU2642302C1 (en) 2009-08-13 2018-01-24 Акселерон Фарма Инк. Combined administration of gdf traps and erythropoetin receptor activators for increasing content of erythrocytes
IN2012DN02766A (en) 2009-09-09 2015-09-18 Acceleron Pharma Inc
ES2869864T3 (en) 2009-11-03 2021-10-26 Acceleron Pharma Inc Procedures for treating fatty liver disease
JP6267425B2 (en) 2009-11-17 2018-01-24 アクセルロン ファーマ, インコーポレイテッド ACTRIIB protein and its variants and uses thereof for utrophin induction for the treatment of muscular dystrophy
KR20130132824A (en) * 2010-11-08 2013-12-05 악셀레론 파마 인코포레이티드 Actriia binding agents and uses thereof
EP2797620B1 (en) 2011-10-17 2019-04-24 Acceleron Pharma Inc. Compositions for treating iron overload in thalassemia
PT2780368T (en) 2011-11-14 2018-03-22 Regeneron Pharma Compositions and methods for increasing muscle mass and muscle strength by specifically antagonizing gdf8 and/or activin a
KR101995751B1 (en) 2012-01-05 2019-07-03 주식회사 와이바이오로직스 Myostatin inhibitor including extracellular domain of delta-like 1 homolog
AU2013274347B2 (en) 2012-06-11 2018-03-08 Amgen Inc. Dual receptor antagonistic antigen-binding proteins and uses thereof
WO2014138485A1 (en) 2013-03-08 2014-09-12 Irm Llc Ex vivo production of platelets from hematopoietic stem cells and the product thereof
MX2015011430A (en) 2013-03-15 2016-04-20 Amgen Inc Myostatin antagonism in human subjects.
AU2014262843B2 (en) 2013-05-06 2017-06-22 Scholar Rock, Inc. Compositions and methods for growth factor modulation
TWI655207B (en) * 2013-07-30 2019-04-01 再生元醫藥公司 Anti-activin A antibody and use thereof
EP3094751A4 (en) 2014-01-14 2017-06-07 Santa Maria Biotherapeutics, Inc. Activin inhibitor response prediction and uses for treatment
AU2015231022B2 (en) 2014-03-21 2021-02-04 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating ineffective erythropoiesis by inhibiting activin B and/or GDF11
JP6649895B2 (en) 2014-04-18 2020-02-19 アクセルロン ファーマ, インコーポレイテッド Method for increasing red blood cell levels and treating sickle cell disease
WO2015192127A2 (en) 2014-06-13 2015-12-17 Santa Maria Biotherapeutics, Inc. Formulated receptor polypeptides and related methods
WO2015192111A1 (en) 2014-06-13 2015-12-17 Acceleron Pharma, Inc. Methods and compositions for treating ulcers
GB201421379D0 (en) 2014-12-02 2015-01-14 Isis Innovation Ltd And Medical Res Council Molecule
HUE062189T2 (en) 2014-12-03 2023-09-28 Celgene Corp Activin-actrii antagonists and uses for treating myelodysplastic syndrome
MA41119A (en) 2014-12-03 2017-10-10 Acceleron Pharma Inc METHODS OF TREATMENT OF MYELODYSPLASIC SYNDROMES AND SIDEROBLASTIC ANEMIA
PL3280727T3 (en) 2015-04-06 2021-07-19 Acceleron Pharma Inc. Single-arm type i and type ii receptor fusion proteins and uses thereof
CN107683287B (en) 2015-04-22 2022-05-06 奥健公司 Novel hybrid ACTRIIB ligand trap proteins for the treatment of muscle wasting diseases
JOP20160092B1 (en) 2015-05-13 2023-03-28 Celgene Corp Treatment of beta-thalassemia using actrii ligand traps
EP4190805A3 (en) 2015-05-20 2023-08-16 Celgene Corporation In vitro cell culture methods for beta-thalassemia using activin type ii receptor ligand traps
EP3370754A4 (en) 2015-11-04 2019-10-23 Acceleron Pharma Inc. Methods for increasing red blood cell levels and treating ineffective erythropoiesis
KR20180096645A (en) 2015-11-23 2018-08-29 악셀레론 파마 인코포레이티드 How to treat eye diseases
KR20180128405A (en) 2016-02-22 2018-12-03 악셀레론 파마 인코포레이티드 ActRII antagonists for use in increasing immune activity
EP3496739B1 (en) 2016-07-15 2021-04-28 Acceleron Pharma Inc. Compositions comprising actriia polypeptides for use in treating pulmonary hypertension
US20180050085A1 (en) 2016-07-27 2018-02-22 Acceleron Pharma Inc. Methods and compositions for treating myelofibrosis
CN110036025B (en) 2016-10-05 2024-03-22 阿塞勒隆制药公司 Variant ActRIIB proteins and uses thereof
US11976111B2 (en) 2016-10-05 2024-05-07 Acceleron Pharma Inc. ActRIIa and ALK4 polypeptides for treating kidney fibrosis, inflammation and injury
JOP20190085A1 (en) 2016-10-20 2019-04-17 Biogen Ma Inc Methods for treating muscle wasting and bone disease using novel hybrid actriib ligand trap proteins
AU2017357931A1 (en) 2016-11-10 2019-06-20 Keros Therapeutics, Inc. GDNF fusion polypeptides and methods of use thereof
CA3043180A1 (en) 2016-11-10 2018-05-17 Keros Therapeutics, Inc. Activin receptor type iia variants and methods of use thereof
GB201620119D0 (en) 2016-11-29 2017-01-11 Pharmafox Therapeutics Ag Compounds
JP7246617B2 (en) 2017-02-01 2023-03-28 アクセルロン ファーマ インコーポレイテッド TGFβ and ACTRII antagonists for use in increasing immune activity
EP3576776A4 (en) 2017-02-06 2020-10-14 Acceleron Pharma Inc. Compositions and methods for treating heart failure
AU2018364668A1 (en) * 2017-11-09 2020-06-11 Keros Therapeutics, Inc. Activin receptor type lla variants and methods of use thereof
EP3737406A4 (en) 2018-01-12 2021-11-03 Keros Therapeutics, Inc. Activin receptor type iib variants and methods of use thereof
KR20210006952A (en) 2018-05-09 2021-01-19 케로스 테라퓨틱스, 인크. Activin receptor type IIA variant and methods of use thereof
WO2021189006A1 (en) 2020-03-20 2021-09-23 Keros Therapeutics, Inc. Methods of using activin receptor type iia variants
IL296394A (en) 2020-03-20 2022-11-01 Keros Therapeutics Inc Activin receptor type ii chimeras and methods of use thereof
EP4121088A1 (en) 2020-03-20 2023-01-25 Keros Therapeutics, Inc. Methods of using activin receptor type iib variants

Also Published As

Publication number Publication date
JP7051846B2 (en) 2022-04-11
US11090361B2 (en) 2021-08-17
EP3538128A2 (en) 2019-09-18
CN110461349A (en) 2019-11-15
US11717558B2 (en) 2023-08-08
AU2017357944A1 (en) 2019-06-20
JP7379573B2 (en) 2023-11-14
US20210275637A1 (en) 2021-09-09
EP3538123A4 (en) 2020-10-14
JP2019535268A (en) 2019-12-12
JP2022088575A (en) 2022-06-14
WO2018089706A3 (en) 2018-09-20
CA3043180A1 (en) 2018-05-17
US20190282663A1 (en) 2019-09-19
EP3538128A4 (en) 2020-07-22
CA3043184A1 (en) 2018-05-17
US11013785B2 (en) 2021-05-25
WO2018089715A1 (en) 2018-05-17
JP7139326B2 (en) 2022-09-20
JP2020503262A (en) 2020-01-30
JP2023181445A (en) 2023-12-21
WO2018089706A2 (en) 2018-05-17
US20190345225A1 (en) 2019-11-14
CN110430890A (en) 2019-11-08
AU2017357935A1 (en) 2019-06-20
AU2017357935B2 (en) 2022-04-28
US20240050528A1 (en) 2024-02-15

Similar Documents

Publication Publication Date Title
US11090361B2 (en) Activin receptor type IIa variants and methods of use thereof
US11484573B2 (en) Activin receptor type IIa variants and methods of use thereof
US20200407415A1 (en) Activin receptor type iib variants and methods of use thereof
IL274735A (en) Compositions and methods related to engineered fc constructs
CA3098679A1 (en) Activin receptor type iia variants and methods of use thereof
WO2022072882A1 (en) Methods of using activin receptor type ii variants
NZ794251A (en) Activin receptor type iia variants and methods of use thereof

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190607

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40014733

Country of ref document: HK

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Free format text: PREVIOUS MAIN CLASS: A61K0038000000

Ipc: C07K0014710000

A4 Supplementary search report drawn up and despatched

Effective date: 20200915

RIC1 Information provided on ipc code assigned before grant

Ipc: C12P 21/02 20060101ALI20200909BHEP

Ipc: C07K 14/71 20060101AFI20200909BHEP

Ipc: A61K 38/17 20060101ALI20200909BHEP

Ipc: C12N 9/12 20060101ALI20200909BHEP

Ipc: A61P 3/00 20060101ALI20200909BHEP

Ipc: A61K 38/00 20060101ALI20200909BHEP

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230518